1

 

                DEPARTMENT OF HEALTH AND HUMAN SERVICES

 

                      FOOD AND DRUG ADMINISTRATION

 

                CENTER FOR DRUG EVALUATION AND RESEARCH

 

 

 

             ADVISORY COMMITTEE FOR PHARMACEUTICAL SCIENCE

 

 

 

 

 

 

 

                        Tuesday, April 13, 2004

 

                               8:30 a.m.

 

 

 

             Advisors and Consultants Staff Conference Room

                           5630 Fishers Lane

                          Rockville, Maryland

 

                                                                 2

 

                              PARTICIPANTS

 

      Arthur H. Kibbe, Ph.D., Chair

      Hilda F. Scharen, M.S., Executive Secretary

 

      MEMBERS

 

                Charles Cooney, Ph.D.

                Patrick P. DeLuca, Ph.D.

                Meryl H. Karol, Ph.D.

                Melvin V. Koch, Ph.D.

                Marvin C. Meyer, Ph.D.

                Gerald P. Migliaccio (Industry

      Representative)

                Cynthia R.D. Selassie, Ph.D.

                Nozer Singpurwalla, Ph.D.

                Marc Swadener, Ed.D. (Consumer

      Representative)

                Jurgen Venitz, M.D., Ph.D.

 

      SPECIAL GOVERNMENT EMPLOYEES

 

                Judy Boehlert, Ph.D.

                Paul H. Fackler, Ph.D., (Acting Industry

                        Representative)

                Thomas P. Layloff, Jr., Ph.D.

 

      FDA

 

                Ajaz Hussain, Ph.D.

                Chris Joneckis, Ph.D.

                Robert O'Neill, Ph.D.

                Keith Webber, Ph.D.

                Helen Winkle

 

                                                                 3

 

                            C O N T E N T S

                                                              PAGE

 

      Call to Order:

                Arthur Kibbe, Ph.D.                              4

 

      Conflict of Interest Statement:

                Hilda Scharen, M.S.                              4

 

      Introduction to Meeting:

                Helen Winkle                                     8

 

      Subcommittee Reports:

                Jurgen Venitz, M.D., Ph.D.                      34

 

      Parametric Tolerance Interval Test

      for Dose Content Uniformity:

                Ajaz Hussain, Ph.D.                             44

 

      Moving Forward--An Approach for Resolution:

                Robert O'Neill, Ph.D.                           46

 

      Committee Discussions and Recommendations                 53

 

      Process Analytic Technology (PAT)--Next Steps:

                Ajaz Hussain, Ph.D.                             70

 

      Finalizing PAT Guidance, Training and

      Certification:

                Chris Watts, Ph.D.                              89

 

      Standards Development:

                Ali Afnan, Ph.D.                               100

 

      Rapid Microbial Methods:

                Bryan Riley, Ph.D.                             110

 

      Committee Discussions and Recommendations                135

 

      Open Public Hearing

                Leo Lucisano, GlaxoSmithKline                  135

                Parrish M. Galliher, Xcellerex                 145

                Troy J. Logan, Siemens                         174

                Robert Mattes, Foss-NIRSystems                 185

 

      PAT Applications for Products in the Office of

      Biotechnology Products (OBP): Overview and Issues:

                Keith Webber, Ph.D.                            194

                Christopher Joneckis, Ph.D.                    213

                Charles Cooney, Ph.D.                          224

                Kevin Koch, Ph.D.                              248

                Tom Layloff, Ph.D.                             271

 

      Committee Discussions and Recommendations                279

 

                                                                 4

 

  1                      P R O C E E D I N G S

 

  2                          Call to Order

 

  3             DR. KIBBE:  Ladies and gentlemen, shall we

 

  4   begin. This is the Advisory Committee for

 

  5   Pharmaceutical Science. Today is April 13th.  Those

 

  6   of you who have not done your taxes, because you

 

  7   are here working for us and the Federal Government,

 

  8   you will get exactly no compensation to allow you

 

  9   to do your taxes late.

 

 10             Hilda.

 

 11                  Conflict of Interest Statement

 

 12             MS. SCHAREN:  Good morning.  I am going to

 

 13   start reading the Conflict of Interest Statement

 

 14   for the Advisory Committee for Pharmaceutical

 

 15   Science.  I am Hilda Scharen with the Center for

 

 16   Drugs, FDA.  I am the Executive Secretary for this

 

 17   committee.

 

 18             The following announcement addresses the

 

 19   issue of conflict of interest with respect to this

 

 20   meeting and is made a part of the record to

 

 21   preclude even the appearance of such at this

 

 22   meeting.

 

 23             Based on the agenda, it has been

 

 24   determined that the topics of today's meeting are

 

 25   issues of broad applicability and there are no

 

                                                                 5

 

  1   products being approved at this meeting.  Unlike

 

  2   issues before a committee in which a particular

 

  3   product is discussed, issues of broader

 

  4   applicability involve many industrial sponsors and

 

  5   academic institutions.

 

  6             All Special Government Employees have been

 

  7   screened for their financial interests as they may

 

  8   apply to the general topics at hand.  To determine

 

  9   if any conflict of interest existed, the Agency has

 

 10   reviewed the agenda and all relevant financial

 

 11   interests reported by the meeting participants.

 

 12             The Food and Drug Administration has

 

 13   granted general matter waivers to the Special

 

 14   Government Employees participating in this meeting

 

 15   who require a waiver under Title 18, United States

 

 16   Code, Section 208.

 

 17             A copy of the waiver statements may be

 

 18   obtained by submitting a written request to the

 

 19   Agency's Freedom of Information Office, Room 12A-30

 

 20   of the Parklawn Building.

 

 21             Because general topics impact so many

 

 22   entities, it is not prudent to recite all potential

 

 23   conflicts of interest as they apply to each member

 

 24   and consultant and guest speaker.

 

 25             FDA acknowledges that there may be

 

                                                                 6

 

  1   potential conflicts of interest, but because of the

 

  2   general nature of the discussion before the

 

  3   committee, these potential conflicts are mitigated.

 

  4             With respect to FDA's invited industry

 

  5   representatives, we would like to disclose that

 

  6   Gerald Migliaccio is participating in this meeting

 

  7   as an industry representative acting on behalf of

 

  8   regulated industry.  Mr. Migliaccio is employed by

 

  9   Pfizer.  Dr. Paul Fackler is participating in this

 

 10   meeting as an acting industry representative.  Dr.

 

 11   Fackler is employed by Teva Pharmaceuticals U.S.A.

 

 12             In the event that the discussions involve

 

 13   any other products or firms not already on the

 

 14   agenda for which FDA participants have a financial

 

 15   interest, the participants' involvement and their

 

 16   exclusion will be noted for the record.

 

 17             With respect to all other participants, we

 

 18   ask in the interest of fairness that they address

 

 19   any current or previous financial involvement with

 

 20   any firm whose product they may wish to comment

 

 21   upon.

 

 22             Thank you.

 

 23             DR. KIBBE:  Thank you.  I am Art Kibbe.  I

 

 24   am Chairman of the Pharmaceutical Science's

 

 25   Department at Wilkes University.

 

                                                                 7

 

  1             We have a tradition of introducing

 

  2   everyone around the table, so, Dr. O'Neill, if you

 

  3   will start.

 

  4             DR. O'NEILL:  I am Bob O'Neill.  I am

 

  5   Director of the Office of Biostatistics in CDER.

 

  6              DR. HUSSAIN:  Ajaz Hussain, Deputy

 

  7   Director, Office of Pharmaceutical Science.

 

  8             MS. WINKLE:  Helen Winkle, Director,

 

  9   Office of Pharmaceutical Science.

 

 10             DR. VENITZ:  Jurgen Venitz, Clinical

 

 11   Pharmacologist, Virginia Commonwealth University.

 

 12             DR. SELASSIE:  Cynthia Selassie, Professor

 

 13   of Chemistry, Pomona College.

 

 14             DR. BOEHLERT:  Judy Boehlert.  I have my

 

 15   own pharmaceutical consulting business.

 

 16             DR. SWADENER:  Marc Swadener, retired from

 

 17   the University of Colorado at Boulder.

 

 18             DR. MEYER:  Marvin Meyer, Emeritus

 

 19   Professor, University of Tennessee, now a

 

 20   consultant in Boca Raton, Florida.

 

 21             DR. KAROL:  Meryl Karol, a Professor at

 

 22   the University of Pittsburgh in Environmental and

 

 23   Occupational Health.

 

 24             DR. LAYLOFF:  Tom Layloff, Management

 

 25   Sciences for Health, a nonprofit, working primarily

 

                                                                 8

 

  1   in Africa on drug quality.

 

  2             DR. KOCH:  Mel Koch, Director of the

 

  3   Center for Process Analytical Chemistry at the

 

  4   University of Washington.

 

  5             DR. COONEY:  Charles Cooney, Department of

 

  6   Chemical Engineering at MIT.

 

  7             DR. DeLUCA:  Pat DeLuca, Professor of

 

  8   Pharmacy at the University of Kentucky.

 

  9             MR. MIGLIACCIO:  Gerry Migliaccio, Vice

 

 10   President of Global Quality Operations for Pfizer.

 

 11             DR. FACKLER:  Paul Fackler, Teva

 

 12   Pharmaceuticals.

 

 13             DR. KIBBE:  Thank you.

 

 14             Next on our agenda is an Introduction to

 

 15   the Meeting.  Ms. Winkle.

 

 16                     Introduction to Meeting

 

 17             MS. WINKLE:  Thank you and good morning to

 

 18   all the committee members.  I especially want to

 

 19   welcome the members who have not attended before or

 

 20   are just joining us for the first time.

 

 21             That includes Dr. Cooney, Dr. Koch, and

 

 22   Dr. Singpurwalla, who is not here yet, but will be

 

 23   joining us later today, and also to Gerry and Paul

 

 24   for helping us out as industry reps.  We are really

 

 25   pleased to have both of them here working with us.

 

                                                                 9

 

  1             [Slide.]

 

  2             Today, I just want to give a short

 

  3   update--it will probably be longer than short, but

 

  4   it is supposed to be short--update on some of the

 

  5   things that we are doing in OPS.

 

  6             [Slide.]

 

  7             Today, I want to talk a little bit about

 

  8   the OPS mission, vision, and goals.  I think it is

 

  9   really important for me to go over these with the

 

 10   committee because it helps all of us understand a

 

 11   little bit more about where OPS is going in the

 

 12   future.  I think that as we talk about various

 

 13   scientific issues, it will help put things in a

 

 14   better perspective for the committee.

 

 15             We have just recently finalized the

 

 16   mission, vision, and goals, so I think it is

 

 17   important that I share them.

 

 18             I also want to talk a little bit about

 

 19   what we are doing in OPS in developing a new

 

 20   paradigm for CMC review in our Office of New Drug

 

 21   Chemistry.  This is really an exciting effort that

 

 22   we have undergone, and I think there are a lot of

 

 23   things that will be very beneficial to talk about a

 

 24   little bit here.

 

 25             A lot of this is built on the

 

                                                                10

 

  1   Pharmaceutical Quality Initiative for the 21st

 

  2   Century, so it helps put that in perspective, as

 

  3   well as to what we are doing in the future in OPS.

 

  4             I also want to mention some of the new

 

  5   personnel that we have in OPS and then talk a few

 

  6   minutes about the meeting agenda.

 

  7             [Slide.]

 

  8             The mission statement.  Again, I think

 

  9   this is very important because it sets forth what

 

 10   OPS is currently focused on, and it is important

 

 11   not only to those activities that we are engaged in

 

 12   and working on very diligently in the organization,

 

 13   they are also very important in supporting the

 

 14   overall mission of the Center and mission of the

 

 15   Agency.

 

 16             Basically, our mission statement is to

 

 17   ensure timely availability of high quality drug

 

 18   products to U.S. patients.  We are doing this

 

 19   through effective and efficient scientific

 

 20   assessment of relevant pharmaceutical and

 

 21   biotechnology information in the submissions, and

 

 22   by facilitating those scientific and technological

 

 23   innovation that improve understanding of product

 

 24   performance, quality, and efficiency of

 

 25   development, manufacturing, and quality assurance

 

                                                                11

 

  1   processes.

 

  2             Many of these things that we have talked

 

  3   about at past meetings, that we will talk about in

 

  4   the future, fall very much within this mission

 

  5   statement and some of the things that we are trying

 

  6   to accomplish.

 

  7             [Slide.]

 

  8             Our vision is to be an international

 

  9   champion.  I think it is very important that we

 

 10   talk about where OPS is going from an international

 

 11   perspective because things are more global.

 

 12             Obviously, now industry, many of the

 

 13   things that we work on are global, and we need to

 

 14   be part of that overall global involvement in

 

 15   pharmaceutical science, but we really want to be

 

 16   champions and leaders in the regulatory application

 

 17   of contemporary scientific knowledge, and that

 

 18   knowledge that affects the design, development,

 

 19   manufacture, and clinical performance of

 

 20   pharmaceutical and biotechnology products.

 

 21             [Slide.]

 

 22             Basically, the goals are for OPS programs

 

 23   and projects to support the achievement of the

 

 24   following attributes of drug products:

 

 25              The drug quality and performance is

 

                                                                12

 

  1   achieved and assured through design of effective

 

  2   and efficient development and manufacturing

 

  3   processes;

 

  4             That regulatory specifications are based

 

  5   on a mechanistic understanding of how product and

 

  6   process factors impact product performance;

 

  7             And that there is continuous "real time"

 

  8   assurance of quality.

 

  9             These are all very important objectives

 

 10   that we are striving toward.

 

 11             [Slide.]

 

 12             Also, OPS will implement a review quality

 

 13   system and procedures throughout the organization

 

 14   that will:

 

 15             Recognize the level of scientific

 

 16   knowledge supporting product applications, process

 

 17   validation, and process capability;

 

 18             Apply a risk-based regulatory scrutiny

 

 19   that will relate to the level of scientific

 

 20   understanding of how formulation and manufacturing

 

 21   process factors affect product performance, and the

 

 22   capability of process control strategies to prevent

 

 23   or mitigate risk of poor product performance.

 

 24             [Slide.]

 

 25             I wanted to talk a few minutes now that I

 

                                                                13

 

  1   have talked about sort of the mission and the

 

  2   goals, and you have a feel for where we are going,

 

  3   I want to talk about some of the changes that we

 

  4   are making.  Specifically, I want to talk about the

 

  5   changes we are making in CMC review.

 

  6             To help set the stage for the future, I

 

  7   wanted to go quickly through the FDA Strategic

 

  8   Action Plan that Dr. McClellan initiated when he

 

  9   came on board, I want to talk about the

 

 10   Pharmaceutical Quality for the 21st century, which

 

 11   is a really important initiative that is taking

 

 12   place in the Agency, and is very important to us as

 

 13   we move ahead in the Office of Pharmaceutical

 

 14   Science and some of the things that we are trying

 

 15   to accomplish.

 

 16             I want to talk just a second about

 

 17   resources in our CMC area, because I think without

 

 18   mentioning the resources and the problems that we

 

 19   have in resources, it is hard to understand why the

 

 20   changes are necessary that need to be made in order

 

 21   to improve on how we do review.

 

 22             Also, I want to talk about a few other

 

 23   influences that have happened since the

 

 24   organization was first established in 1995.

 

 25             [Slide.]

 

                                                                14

 

  1             The FDA Strategic Plan - Responding to

 

  2   Challenges and Opportunities.  Again, as I said,

 

  3   Dr. McClellan introduced this plan several months

 

  4   after he entered the Agency.  He was very focused

 

  5   while he was here at the Agency on accomplishing

 

  6   these particular aspects of all of the products

 

  7   that are regulated by FDA.

 

  8             Mainly, he focused on efficient risk

 

  9   management, so that we were sure we were going to

 

 10   get the most public health bang for our regulatory

 

 11   buck.

 

 12             He wanted empowering consumers.  He felt

 

 13   that I think all of us understand there is a lot of

 

 14   interest on the part of consumers in their own

 

 15   health care, and he wanted to be able to improve

 

 16   health through better information to consumers, so

 

 17   as they make decisions, as they look at their own

 

 18   health care, as they even deal with their

 

 19   physicians, et cetera, that they have a better

 

 20   understanding of the medications, food, et cetera,

 

 21   et cetera, that they need to take or use.

 

 22             He wanted to improve patient and consumer

 

 23   safety, protect America from terrorism, and more

 

 24   effective regulation through a stronger workforce.

 

 25             So, as we make changes in OPS, and we look

 

                                                                15

 

  1   toward the future of things that we want to do

 

  2   differently, and how we want to do those, we are

 

  3   trying to incorporate many of the things that Dr.

 

  4   McClellan incorporated in his strategic plan.

 

  5             [Slide.]

 

  6             Also, as I mentioned, the FDA Initiative

 

  7   on Pharmaceutical Quality is an important

 

  8   groundwork for some of the things that we are doing

 

  9   now and in the future in OPS.

 

 10             This particular chart is very helpful

 

 11   because it shows the particular dimensions of the

 

 12   plan for strong public health protection, for

 

 13   international cooperation, for risk-based

 

 14   orientation, science-based policy and standards,

 

 15   and integrated quality systems orientation.  These

 

 16   are the really important aspects of the initiative

 

 17   and where we are going.

 

 18             [Slide.]

 

 19             There are various directional vectors that

 

 20   came with the initiative, and I won't go through

 

 21   each of these.  I think you can look through them,

 

 22   but I think they are important as we look at OPS

 

 23   and where we are going for OPS in the future, so

 

 24   looking at our regulatory policies, making sure

 

 25   that we incorporate new technology advances when we

 

                                                                16

 

  1   do our regulation, that we are able to work with

 

  2   industry, et cetera, in doing some of these, and

 

  3   that we have consistency and coordination

 

  4   throughout the whole drug quality regulatory

 

  5   program.

 

  6             [Slide.]

 

  7             Here is basically the directional vectors

 

  8   and many of the things that are being worked on

 

  9   under the GMP initiative agencywide.

 

 10             These include looking at a preapproval

 

 11   inspection compliance program, dispute resolution

 

 12   processes being established, a pharmaceutical

 

 13   inspectorate that focuses specifically on

 

 14   pharmaceutical products during the inspection

 

 15   process that is being set up.  We are hoping to

 

 16   have product specialists on inspection process, and

 

 17   we hope to start that very soon.

 

 18             We have set guidance on CFR Part 11,

 

 19   aseptic processing guidance, a comparability

 

 20   protocol guidance.  We have been doing a lot of

 

 21   stuff with risk management and quality by design,

 

 22   and, of course, PAT, which we have talked about.

 

 23   But you can see where each of these sits on the

 

 24   whole vector between risk and science.  These are

 

 25   all important aspects of the initiative.

 

                                                                17

 

  1             [Slide.]

 

  2             But the most important thing to me about

 

  3   the initiative is it afforded us in OPS, a lot of

 

  4   opportunities to change the way that we do

 

  5   business.  It has opened up a window of time for us

 

  6   to really look at how we do business and make the

 

  7   changes that are necessary to move forward into the

 

  8   21st century.

 

  9             This is not easy, and I will go through

 

 10   some of the challenges what we have had, but first

 

 11   of all, I want to talk about some of these

 

 12   opportunities and just mention them to you, because

 

 13   I think they are really important.

 

 14             We really have the opportunity now to

 

 15   strategize more on how we are going to ensure

 

 16   product quality.  This is ensuring product quality

 

 17   across all of the Center, and it is the first time

 

 18   we really have thought about the whole aspect of

 

 19   product quality and what needs to be done to ensure

 

 20   in  the future that we are focused on the right

 

 21   aspects of that.

 

 22             We need to revisit our processes.  This is

 

 23   a really good opportunity for us to do that.  We

 

 24   have built processes over the last 20, 25 years,

 

 25   not only in review, but in inspection, as well, and

 

                                                                18

 

  1   this gives us an opportunity to look at all of the

 

  2   processes that fall under pharmaceutical quality

 

  3   regulation, and to incorporate best practices.

 

  4             We need to focus more on manufacturing and

 

  5   associated issues relating to the quality of

 

  6   products, one of the things that was very apparent

 

  7   to us when we went in and looked at the review

 

  8   processes, that we did not pay as much attention to

 

  9   the actual manufacturing of products and how it

 

 10   affected the quality of the products.

 

 11             So, this is a really good opportunity for

 

 12   us to do that.  We have a lot to learn and we have

 

 13   to work with a lot of people because obviously, we

 

 14   don't have as much understanding as we need, but we

 

 15   are doing a lot and looking at manufacturing

 

 16   science and trying to get a better understanding of

 

 17   that.  I think that has been very apparent in some

 

 18   of the things that you have talked about with PAT,

 

 19   we will talk about even more today.

 

 20             We need to focus both on review and

 

 21   inspection, and we need to put more science into

 

 22   those.  A lot of times, and it has been said time

 

 23   and time again, we have not used really good

 

 24   science in making the decision, and sometimes we

 

 25   have had a lot of complaints from industry and

 

                                                                19

 

  1   others about that lack of really scientific

 

  2   understanding on inspections, so this is a really

 

  3   good opportunity for us to ensure that that science

 

  4   exists, but it is really important that we ensure

 

  5   that it is part of the review process, as well, and

 

  6   it is going to take time, but I think working with

 

  7   our people and others, we will get there in the

 

  8   future.

 

  9             We need to enhance the interactions

 

 10   between review, inspection, and compliance.  One of

 

 11   the things that was very interesting to me right

 

 12   before we started the initiative is we met with a

 

 13   number of people from trade associations, and it

 

 14   was made very clear, the gap between what happens

 

 15   in review and what happens in inspection, and who

 

 16   is sitting in the middle but industry with a lot of

 

 17   questions on how policy was set or what the policy

 

 18   means, and dealing with the inspectors day to day

 

 19   who really don't have an understanding of that

 

 20   either, so we really need to ensure better

 

 21   interaction between review and inspection.

 

 22             We need to foster communication with

 

 23   industry.  In the review, we have been very

 

 24   hesitant to talk much to the industry and to work

 

 25   with the industry, not only on specific

 

                                                                20

 

  1   applications, but on science in general, and there

 

  2   is a lot of science in the industry that can be

 

  3   very beneficial to us in the Agency to understand

 

  4   the processes and understand manufacturing and

 

  5   pharmaceutical quality, and we need to do more of

 

  6   that.

 

  7             We need to have early discussion on CMC

 

  8   questions. As I already mentioned, we have a

 

  9   dispute resolution process that we are setting up,

 

 10   which we feel will be very helpful to give industry

 

 11   an opportunity to talk with us when they have

 

 12   scientific issues or questions.

 

 13             We need to leverage resources for the best

 

 14   bang for the buck.  This is a real problem, and as

 

 15   I said, I am going to talk a little bit more about

 

 16   resources.

 

 17             We need to simplify the regulatory

 

 18   requirements and we need to be able to find ways to

 

 19   reduce some of the regulatory burden.  We have

 

 20   talked here before at the committee about the

 

 21   number of supplements that we get in the

 

 22   organization, we are really drowning in

 

 23   applications in supplements, and all of them are

 

 24   treated basically the same, and we need to really

 

 25   step back and look at ways that we can put more

 

                                                                21

 

  1   emphasis or more responsibility on industry and try

 

  2   and work with them to have better understanding of

 

  3   things, and not get as many applications.

 

  4             We need to eliminate the "check box"

 

  5   approach that we have.  What we do basically in

 

  6   review is we go through and do you have this, do

 

  7   you have that, do you have this without a real

 

  8   understanding of what the process is, the

 

  9   manufacturing, the whole important aspects of

 

 10   pharmaceutical quality.

 

 11             [Slide.]

 

 12             We need to enhance training opportunities,

 

 13   and we now have this opportunity under the GMP

 

 14   initiative, as well as some of the things that we

 

 15   are undertaking in OPS.  We are in the process of

 

 16   working with several of the pharmaceutical

 

 17   industries to set up plant residency programs for

 

 18   some of our chemists.

 

 19             We have other cross-training opportunities

 

 20   that we are discussing, and then we have the

 

 21   pharmaceutical inspectorate, and the reason I put

 

 22   this here is not only will we be able to train our

 

 23   inspectors better as far as some of the aspects or

 

 24   manufacturing science, will it be able to take

 

 25   advantage of those from the review standpoint, as

 

                                                                22

 

  1   well, and I think this will be extremely helpful

 

  2   and useful to us in our future regulatory

 

  3   activities.

 

  4             [Slide.]

 

  5             We need to enhance FDA's knowledge

 

  6   regarding new technologies in manufacturing, and we

 

  7   need to encourage innovation, and again this goes

 

  8   back to PAT.

 

  9              We need to develop processes that are

 

 10   focused more on product risk, which we have not

 

 11   done.  As I said before, almost every product has

 

 12   the same weight, same level of review, and we

 

 13   really need to look more at the risk aspects of the

 

 14   product.

 

 15             We need to revisit how quality of products

 

 16   relate to ensuring safety and efficacy, and

 

 17   especially ensuring clinical relevance.

 

 18             We need to alleviate industry's concern

 

 19   regarding reprisal.  I hate to put this up, it's a

 

 20   bad word "reprisal," but that thought is out there

 

 21   often in industry, I hear it time and time again,

 

 22   and I am hoping through better interactions with

 

 23   industry, with better understanding of the science

 

 24   and the ability to discuss the science, we can

 

 25   begin to eliminate some of these concerns.

 

                                                                23

 

  1             We need to enhance our international

 

  2   involvement. We are working on pharmaceutical

 

  3   development and risk management in international,

 

  4   but we need to do more of this, because again it's

 

  5   a very global world out there, and we need to be

 

  6   sure that we are involved in everything that is

 

  7   happening on the international front.

 

  8             [Slide.]

 

  9             I did say I wanted to mention resources

 

 10   real quickly.  I thought this would give you a

 

 11   better perspective again as to why we want to make

 

 12   some of the changes in CMC. The workload is really

 

 13   difficult for our CMC reviewers in new drugs.

 

 14              We got, in 2003, 159 NDAs, 342 commercial

 

 15   INDs, 507 research INDs, 1,858 CMC supplements, and

 

 16   that doesn't include efficacy or labeling

 

 17   supplements, and 1,132 annual reports.  This is a

 

 18   lot of work to take on, and this is a lot of work

 

 19   because we have fewer and fewer review staff.

 

 20             We have constantly been over the last few

 

 21   years hit by reductions in resources, so we are

 

 22   doing more work with less people, and we have

 

 23   really got to think of ways to streamline the

 

 24   process and to be able to get some of this done in

 

 25   a more efficient and effective manner.

 

                                                                24

 

  1             [Slide.]

 

  2             Other influences, though, too, that bring

 

  3   about the necessity for change, as I said, in 1995,

 

  4   when ONDC was established, it was collocated with

 

  5   the clinical divisions, and this seemed to work

 

  6   really well for a couple of years, but a lot of

 

  7   things have happened within the Center, within the

 

  8   Agency, within the world, that really affect how we

 

  9   do the CMC reviews, so we really need to rethink,

 

 10   based on these influences and changes, how we do

 

 11   things.

 

 12             Some of the influences includes shorter

 

 13   PDUFA deadlines, FDAMA, again harmonization and

 

 14   globalization, such changes in our regulatory

 

 15   processes, such as SUPAC, BACPAC, new technologies

 

 16   in pharmaceutical manufacturing.

 

 17             [Slide.]

 

 18             PAT, counterterrorism, counterfeit

 

 19   products.  We were just talking about the fact that

 

 20   we can't even begin to keep up with counterfeiting,

 

 21   we have to find better ways to do that.

 

 22             BSE and other crisis, such as that.  There

 

 23   has been a greater focus on generic drugs, and

 

 24   tomorrow we will spend a lot of time talking about

 

 25   some of the issues that we have with regulating

 

                                                                25

 

  1   generic products, and it is really important that

 

  2   we begin to focus more on some of these issues and

 

  3   how we need to ensure that we incorporate other

 

  4   thinking from the new drug side into how we are

 

  5   going to regulate generic products in the future.

 

  6             There have been a lot of changes in

 

  7   industry, more globalization mergers, et cetera.

 

  8   There has been electronic submissions.  We are

 

  9   working very hard to hopefully enhance the

 

 10   efficiency of our processes through electronic

 

 11   submissions, and there has been more focus on risk

 

 12   management and quality systems.

 

 13             [Slide.]

 

 14             So, basically, what we need to do is to

 

 15   change the paradigm for CMC review.  I have talked

 

 16   about that we have the opportunity to do this.  The

 

 17   things that we really need to focus on based on

 

 18   those opportunities is really to develop a

 

 19   risk-based CMC review.

 

 20             I think this is really important, and I

 

 21   think we are going to need help.  This is not going

 

 22   to be an easy thing to determine risk.

 

 23             I think products are going to come and go

 

 24   that are risky, we see that all the time, products

 

 25   that you don't expect when it comes on the market

 

                                                                26

 

  1   to have any risk, then, things are found out later

 

  2   on, so it is not going to be an easy process to

 

  3   develop, and it is going to take a lot of thought

 

  4   and probably a lot of help even from the committee,

 

  5   but this is definitely a direction that we need to

 

  6   head in.

 

  7             We need to establish quality systems which

 

  8   help set the framework for ensuring that we do have

 

  9   a dynamic organization and that we can handle the

 

 10   complications of the regulatory processes.

 

 11             We need to focus resources towards efforts

 

 12   that improve quality, and not hinder and interfere

 

 13   with innovation, and I think that is very

 

 14   important, and we need to focus on all aspects of

 

 15   CMC.

 

 16             We need to look at chemistry, we need to

 

 17   look at manufacturing, and we need to look at

 

 18   controls, and we have not done as good a job of

 

 19   this in the past.

 

 20             [Slide.]

 

 21             The advantages of the new paradigm, for

 

 22   FDA, we will have more product and process

 

 23   knowledge, which can be shared by industry, so that

 

 24   we have a better understanding of the products that

 

 25   we regulate.

 

                                                                27

 

  1              We will have more efficient resource

 

  2   allocation for review and inspection, and we can

 

  3   increase our trust and understanding  of industry

 

  4   decision making.

 

  5             [Slide.]

 

  6             The advantages for industry is hopefully,

 

  7   that we will have fewer, more efficient,

 

  8   science-based inspections, faster, more consistent

 

  9   reviews.

 

 10             There is a potential for reduced

 

 11   regulatory burden, for managing changes with less

 

 12   FDA oversight, for focused resources on critical

 

 13   issues, flexibility to focus on what should be

 

 14   done, not what can be done, and to improve

 

 15   communication with FDA.

 

 16             [Slide.]

 

 17             But most of all, the ultimate beneficiary

 

 18   is the public, and we hope through some of the

 

 19   changes that we make, that we can increase the

 

 20   availability of drugs on the market, we can have

 

 21   faster approval of new products, we can have

 

 22   continued assurance of high quality products, and

 

 23   we can increase the public's confidence in the work

 

 24   that we are doing in FDA, and hopefully, reduce

 

 25   costs, which isn't, of course, our business, but

 

                                                                28

 

  1   something we hope is going to come out of some of

 

  2   the changes that we are making.

 

  3             [Slide.]

 

  4             The new paradigm will include developing

 

  5   strategies to recruit and train reviewers.  One of

 

  6   the things that we realize is that we have a real

 

  7   gap in the qualifications that our reviewers have.

 

  8             We need more that have understanding of

 

  9   drug discovery, analytical chemistry,

 

 10   pharmaceutical engineering, and we are going to be

 

 11   looking at recruiting and training people in these

 

 12   areas.

 

 13             We need to build a learning organization,

 

 14   one that is skilled at creating, acquiring, and

 

 15   transferring knowledge.  This is one thing we have

 

 16   not done an adequate job of in the past, and we

 

 17   really need to work on, probably not only just in

 

 18   OPS, but throughout the whole Center.

 

 19             We need to set specifications based on

 

 20   science and process understanding.  We need to

 

 21   reengineer the process, so that we have the best

 

 22   practices, metrics, and that we are customer

 

 23   oriented.

 

 24             This is another thing that we have not

 

 25   paid a lot of attention to in the past, which we

 

                                                                29

 

  1   really need to look toward in the future, is who

 

  2   our customers are and what they need.

 

  3             [Slide.]

 

  4             We need to increase emphasis on

 

  5   manufacturing science, we need to ask the right

 

  6   questions at the right time.  We need to implement

 

  7   peer review by FDA scientists and clinicians.

 

  8             Establish a program to better integrate

 

  9   review and inspection, develop processes which

 

 10   ensure regulatory relief based on process

 

 11   understanding and control, quality systems in

 

 12   manufacturing, and continuous improvement is very

 

 13   important, and we need to create a better work

 

 14   environment and promote job satisfaction within our

 

 15   organization.

 

 16             [Slide.]

 

 17             As I said, there is a lot of challenges.

 

 18   The current culture, both inside and outside of

 

 19   FDA, is definitely the biggest challenge we have.

 

 20   It is very difficult to get people to think

 

 21   differently.  They have worked in a certain culture

 

 22   for years and years, and changing that culture is

 

 23   not easy.  We see that both inside the Agency, as

 

 24   well as outside.

 

 25             Hiring is not easy, it is very difficult

 

                                                                30

 

  1   to find people with the right skills that want to

 

  2   come to work for the Government, and this is a big

 

  3   challenge that we have ahead of us.

 

  4             Establishing performance metrics is also a

 

  5   challenge because we have really never had the

 

  6   metrics to measure anything except for the amount

 

  7   of work we get, and we are really going to have to

 

  8   step back and look at this differently.

 

  9             We need to identify gaps in requirements.

 

 10   We need to reevaluate the review process again to

 

 11   be sure we are asking the right questions that

 

 12   ensure product quality.

 

 13             We need to understand what is relevant

 

 14   science.

 

 15             We need to determine what is needed for

 

 16   pharmaceutical development data to assist in a

 

 17   better understanding of manufacturing process.

 

 18             We need to develop a science-based risk

 

 19   model,  and we need to integrate better into the

 

 20   inspection process  including participating on

 

 21   inspections.

 

 22             This is a lot of work we have ahead of us,

 

 23   and the reason I am sharing it is because I think a

 

 24   lot of these issues are going to come up in the

 

 25   future where we are going to need the committee's

 

                                                                31

 

  1   input on how to tackle some of these challenges,

 

  2   some of the things that we need to incorporate into

 

  3   our review and our processes to make sure that we

 

  4   are doing what is necessary to have the best

 

  5   regulatory processes available.

 

  6             Again, I feel that this is important that

 

  7   you all have an understanding of where we are

 

  8   going, and we will look forward to talking about

 

  9   many of these things in the future.

 

 10             [Slide.]

 

 11             Before I go into the agenda, I just wanted

 

 12   to mention some of OPS's new additions that we

 

 13   have.  We are really fortunate to be acquiring a

 

 14   lot of new staff lately, and some of the people I

 

 15   think that are very important, that will be working

 

 16   with us very closely, I wanted to talk about today.

 

 17             First, is Dr. Vince Lee.  I think all of

 

 18   you know Dr. Lee since he was once chair of this

 

 19   committee.  We are very happy to have Vince with

 

 20   us, and we feel that there is a lot of things that

 

 21   he is going to be able to help us work on as we

 

 22   move towards changing some of our regulatory

 

 23   paradigms.

 

 24             Also, we will be adding Dr. Mansor Khan

 

 25   from Texas to our staff.  He is going to be our

 

                                                                32

 

  1   director of our Division for Product Quality

 

  2   Research in our Office of Testing and Research, and

 

  3   he will be joining us next month.

 

  4             We are looking forward, too, to having Dr.

 

  5   Khan.  I think he is going to add a lot and help us

 

  6   a lot in some of the areas of research that we need

 

  7   to be focused on in order to accomplish some of the

 

  8   things that we want to accomplish.

 

  9             Also, I wanted to mention that Dr. Moheb

 

 10   Nasr has become the permanent director of the

 

 11   Office of New Drug Chemistry.  I think many of you

 

 12   know Dr. Chiu has retired. Dr. Nasr so kindly came

 

 13   from St. Louis to take this job, and has been

 

 14   working very diligently on some of the changes that

 

 15   we are trying to make.

 

 16             Dr. Chi Wan Chen has joined him as the

 

 17   deputy of the office.

 

 18             Also, I wanted to announce that Dr. Keith

 

 19   Webber, who is sitting over here, too, is the

 

 20   Acting Director of the Office of Biotech Products.

 

 21   We appreciate Dr. Webber stepping in and taking on

 

 22   this very challenging group that has recently

 

 23   joined us in the Office of Pharmaceutical Science.

 

 24             [Slide.]

 

 25             Just to finalize my presentation, I just

 

                                                                33

 

  1   wanted to quickly go through the meeting topics.  I

 

  2   think this is going to be an extremely exciting

 

  3   meeting.  I think that the topics tomorrow are

 

  4   especially stimulating, topics that I think will

 

  5   add a lot to our future thinking in these areas.

 

  6             Today, we are going to have subcommittee

 

  7   reports.   We are going to have a discussion of the

 

  8   proposal on PTIT.  That is parametric tolerance

 

  9   interval test for dose content uniformity.  We have

 

 10   talked about this before.  We have a proposal now

 

 11   on how we want to finalize our thinking in this

 

 12   area.

 

 13             Then, we want to talk about PAT.  We want

 

 14   to give an update, talk about some of the things

 

 15   that we have done, and also talk about how PAT is

 

 16   going to be implemented in our Office of Biotech

 

 17   Products.

 

 18             Tomorrow, as I said, I think the topics

 

 19   are very stimulating, I think we will have some

 

 20   really good discussion on bioequivalence topics.

 

 21   We want to talk about highly variable drugs, about

 

 22   bioINequivalence.  This is very important.

 

 23             We have a lot of areas here of thought

 

 24   that we need to bring forward and discuss with the

 

 25   committee, and we want to talk about topical

 

                                                                34

 

  1   products.

 

  2             Also, time allowing tomorrow, we have an

 

  3   awareness topic, and this is nanotechnology that we

 

  4   want to introduce.

 

  5             With that, I am going to finish up and

 

  6   hand it over to Dr. Kibbe, and I look forward to

 

  7   hearing the discussion in the next two days.

 

  8             Thank you.

 

  9             DR. KIBBE:  Thank you, Helen.

 

 10             We are pretty close to being on time, so

 

 11   we will turn it over now to the subcommittee

 

 12   reports.  The first one is from Clinical

 

 13   Pharmacology.  Jurgen is moving rapidly to the

 

 14   podium, so here we go.

 

 15                       Subcommittee Reports

 

 16             DR. VENITZ:  Good morning.  I am here to

 

 17   report back from a meeting that the Clinical

 

 18   Pharmacology Subcommittee had last November.

 

 19             [Slide.]

 

 20             Just in terms of review, this committee is

 

 21   serving to provide expertise in three different

 

 22   areas to this parent committee:  pharmacometrics or

 

 23   exposure-response modeling, pediatrics, and

 

 24   pharmacogenetics.  As you see, those were the three

 

 25   topics that we discussed.

 

                                                                35

 

  1             [Slide.]

 

  2             Our first topic in the November meeting

 

  3   was a proposal by Dr. Lesko from OCPB to institute

 

  4   End of Phase 2a Meetings.  Those are meetings that

 

  5   are currently not recommended or that are currently

 

  6   not required by the FDA.

 

  7             He, as well as Dr. Lee, presented the

 

  8   FDA's perspective, and then we had three FDA

 

  9   staffers giving us case reports where those

 

 10   meetings may be helpful in finding optimal doses

 

 11   early on and identifying key issues.

 

 12             [Slide.]

 

 13             The committee appreciated that this was a

 

 14   pilot program that is intended to improve dose

 

 15   findings over a few years.  There was some

 

 16   discussion as to how we assess the success of a

 

 17   program.

 

 18             The committee noticed that there would be

 

 19   additional FDA resources required to implement this

 

 20   very program, but on the positive end, that this

 

 21   End of Phase 2 Meeting Program would allow

 

 22   integration of preclinical information both in the

 

 23   PK and PD area and particularly to identify early

 

 24   on the use of biomarkers in Phase 2 and Phase 3

 

 25   studies that may help streamline the dose finding

 

                                                                36

 

  1   process.

 

  2             The committee also felt that a meeting

 

  3   such as this would be very useful in identifying

 

  4   key issues early on and discuss them between the

 

  5   sponsor and the FDA, as well as define what we call

 

  6   "utility" functions, which are basically measures

 

  7   of the potential consequences of either safety or

 

  8   efficacy issues which are essential to come up with

 

  9   an optimal dose.

 

 10             There was, as I said before, some

 

 11   discussion as to how you would measure the success

 

 12   of such a program, and the committee felt that

 

 13   probably the overriding metrics to measure the

 

 14   success would be customer satisfaction, the

 

 15   customer being both the sponsor, as well as the

 

 16   FDA.

 

 17             Possible, but more difficult to measure

 

 18   outcome would be the need to have post-approval

 

 19   dose changes.  Again, if we can minimize that, that

 

 20   would indicate that there is success in this

 

 21   program.

 

 22             So, while the committee was in support of

 

 23   this program, and as far as I know, it is being

 

 24   implemented as speak.

 

 25             [Slide.]

 

                                                                37

 

  1             The second issue relating to

 

  2   exposure-response was the issue about clinical

 

  3   trial simulations specifically with the intent to

 

  4   assess the liability of drug products to induce QT

 

  5   changes which are thought to be associated with

 

  6   fatal cardiac arrhythmias, we had Dr. Lee give the

 

  7   introduction, Dr. Bonate from the outside review

 

  8   modeling that he had done, clinical trial

 

  9   simulations, and then Dr. Kenna from the FDA review

 

 10   ongoing project within the FDA.

 

 11             [Slide.]

 

 12             There was a lively discussion on this very

 

 13   topic. The committee I think still felt that the

 

 14   QTc correction methods, those are ways to correct

 

 15   the QT interval for change in heart rate, that

 

 16   those methods are still questionable, we still

 

 17   don't have a gold standard on that.

 

 18             We felt that despite the trial simulations

 

 19   presented to us, it still appears very difficult to

 

 20   separate drug-induced changes from baseline changes

 

 21   in those EKG intervals.

 

 22             There was some discussion as to what

 

 23   constitutes a meaningful QTc change.  Right now the

 

 24   perception is that a 6-millisecond average QTc

 

 25   change would be relevant.  There is some concern in

 

                                                                38

 

  1   the committee or there was some concern stated in

 

  2   the committee that that might be too conservative,

 

  3   however, there was acknowledgment that using

 

  4   clinical trial simulation to get to the issue as to

 

  5   what the QTc liability is of a new product may

 

  6   provide a more rational risk/benefit assessment.

 

  7             One issue that was brought up that is

 

  8   currently not being explored is the fact that some

 

  9   drugs, not only interact at the kinetic level, but

 

 10   also the dynamic level, which may lead to QTc

 

 11   changes on the PD level.

 

 12             [Slide.]

 

 13             The second major topic related to the

 

 14   pediatrics component of the committee, here, we

 

 15   reviewed the pediatric decision trees.  We had

 

 16   several speakers.  We had Dr. Hinderling and Dr.

 

 17   Chen giving case reports.  Those were drugs or drug

 

 18   products that were reviewed for the pediatric use,

 

 19   used what is a called a "pediatric decision tree,"

 

 20   that allows PK or PK/PD studies to support efficacy

 

 21   and safety.

 

 22             We had Dr. Machado giving a statistical

 

 23   overview on what methods might be useful to compare

 

 24   pediatric exposure-response to see whether there

 

 25   are any age-related differences.

 

                                                                39

 

  1             Then, our committee member Dr. Kearns gave

 

  2   his perspective on how those studies actually are

 

  3   being done in practice and what some of the

 

  4   shortcomings are of the current pediatric decision

 

  5   tree, and this was followed by Dr. Rodriguez giving

 

  6   the FDA experience with the decision tree that has

 

  7   been in place for a few years.

 

  8             [Slide.]

 

  9             There was some discussion about the age

 

 10   appropriateness of some of the endpoints that are

 

 11   currently required to measure the pharmacology of

 

 12   drugs in children, whether the endpoints are

 

 13   related to the mechanism of action of the drug

 

 14   and/or the pathophysiology of the disease, are

 

 15   those meaningful endpoints and what do they tell

 

 16   us.

 

 17             There was some discussion, because that is

 

 18   part of the decision tree, as to what evidence

 

 19   supports that the disease progression in children

 

 20   is similar to the one in adults, which would then

 

 21   allow it to transfer information from adults to

 

 22   children.

 

 23             There seemed to be consensus that

 

 24   nonclinical information, such as data from primate

 

 25   studies or in-vitro studies may be very useful in

 

                                                                40

 

  1   supporting the pediatric decision tree.

 

  2             However, there was extensive discussion on

 

  3   whether there has to be extensive interaction and

 

  4   discussion between both the clinical pharmacology,

 

  5   the OCPB, as well as the reviewing divisions on the

 

  6   pediatric decision tree and its use in a particular

 

  7   drug product area.

 

  8             There was some discussion also on the

 

  9   limitations of the exposure-response in terms of

 

 10   some of the PD differences that are very difficult

 

 11   to be captured in the current paradigm.

 

 12             I think there was overall an appreciation

 

 13   that the pediatric decision tree is still

 

 14   work-in-progress and additional updates may be

 

 15   necessary to review or start discussing any changes

 

 16   to it.

 

 17             [Slide.]

 

 18             The last area that we discussed related to

 

 19   the pharmacogenomics and the metabolic drug

 

 20   interaction area, so we had two outside speakers,

 

 21   Dr. Flockhart and Dr. Neuvonen talk about two

 

 22   relatively novel cytochrome p450 isoenzymes that

 

 23   start to emerge as part of drug metabolizing

 

 24   enzymes, and the issue was here what is the current

 

 25   state-of-the-art, what can FDA use as basis of

 

                                                                41

 

  1   review for new incoming NDAs.

 

  2             [Slide.]

 

  3             There was acceptance by the committee for

 

  4   cytochrome P4502B6, that we do have both in-vitro,

 

  5   as well as in-vivo, substrates, model substrates

 

  6   that can be used for drug interactions.

 

  7             We don't have, on the other hand, any

 

  8   specific clinical inhibitors, and somewhat

 

  9   questionable in-vitro inhibitors.  On the other

 

 10   hand, for cytochrome P4502C8, we do have both

 

 11   in-vitro, as well as in-vivo, inhibitors, as well

 

 12   as substrates, so we can characterize any

 

 13   interaction potential for cytochrome P4502C8.

 

 14             Discussion by the committee followed that

 

 15   went beyond the specific isoenzymes where the

 

 16   committee emphasized that it is becoming more and

 

 17   more essential to look at population-based clinical

 

 18   studies to primarily assess, not the incidence of

 

 19   drug interactions, but their clinical significance.

 

 20             In other words, we have enough science to

 

 21   support the likelihood of drug-drug interactions,

 

 22   but we are not always sure about what the clinical

 

 23   consequence would be or consequence would be.

 

 24             Along the same line, the committee made

 

 25   the recommendation to encourage sponsors to review

 

                                                                42

 

  1   databases that exist, medication-use databases, to

 

  2   look for this very issue, what are the clinical

 

  3   consequences of drug-drug interactions especially

 

  4   if you go beyond two interactions.

 

  5             [Slide.]

 

  6             The last topic that we discussed related

 

  7   to pharmacogenomics.  Again, this is an ongoing

 

  8   discussion that we had.  In this case, we were

 

  9   discussing how to integrate that in the drug

 

 10   development and what kind of labeling may be

 

 11   necessary to reflect information collected during

 

 12   the development process.

 

 13             We had committee member Dr. Flockhart and

 

 14   Dr. Relling give their academic, as well as

 

 15   clinical, perspective, and Dr. Hockett give the

 

 16   industry perspective.

 

 17             [Slide.]

 

 18             To summarize the committee discussion, I

 

 19   think there was acceptance of the fact that we need

 

 20   additional population-based studies meaning

 

 21   large-scale studies to look at the prevalence for

 

 22   some of the rare genetic polymorphisms, in other

 

 23   words, for some of those polymorphisms that may be

 

 24   important, we do not know how many patients have

 

 25   those specific genotypes.

 

                                                                43

 

  1             There was recognition that we do have or

 

  2   at least start to emerge having a lot of

 

  3   mechanistic and quantitative understanding that is

 

  4   necessary for labeling.

 

  5             In other words, we collect a lot of

 

  6   information and we know a lot about how likely some

 

  7   of those pharmacogenetic differences are and what

 

  8   the kinetic or dynamic consequences are.

 

  9             The discussion then really focused on what

 

 10   is the impact as far as risk/benefit is concerned,

 

 11   in other words, how do we translate changes in drug

 

 12   levels or change in the pharmacology of the drug,

 

 13   how do we translate that into safety and efficacy

 

 14   information.

 

 15             There was, shall we say, a lively

 

 16   discussion of how to label pharmacogenetic

 

 17   information in drug package insert, and I don't

 

 18   think there was any consensus.

 

 19             We had experts telling us we need to label

 

 20   very extensively, on the other hand, clinicians

 

 21   were concerned about overloading information that

 

 22   is not being used by the ultimate consumer, and

 

 23   there was recognition that pharmacogenetics or

 

 24   pharmacogenomics is going to be different from some

 

 25   of the other clinical covariates in the sense that

 

                                                                44

 

  1   it has multidimensional nature, in other words,

 

  2   there are lots of different pharmacogenetic

 

  3   polymorphisms that may be relevant for a given drug

 

  4   product.

 

  5             I would be happy to entertain any

 

  6   questions that you may have.

 

  7             DR. KIBBE:  Okay.  Jurgen will be with us,

 

  8   so if you want to ask questions later, if topics

 

  9   come up that we need to get back to him on, we can.

 

 10             Thank you.

 

 11             Now, I know you are fumbling through your

 

 12   things looking for the slides for the next speaker,

 

 13   but there aren't any, which gives us great hope

 

 14   that it will be a short and direct presentation.

 

 15             Dr. Hussain.

 

 16              Parametric Tolerance Interval Test for

 

 17                     Dose Content Uniformity

 

 18             DR. HUSSAIN:  No, I do not have slides for

 

 19   this part of my introduction.  The topic that will

 

 20   be discussed as a proposal to you is that of

 

 21   parametric tolerance interval test.

 

 22             As we have discussed this several times

 

 23   with you, in particular at the last meeting, in the

 

 24   previous meeting that we had, the challenge is how

 

 25   do you move forward with adopting a more rigorous

 

                                                                45

 

  1   scientific, statistically sound approach to dose

 

  2   content uniformity of inhaled products.

 

  3             We believe that parametric tolerance

 

  4   interval test that is being proposed by IPAC-RS is

 

  5   an improvement over the current method, and we

 

  6   would like to sort of move forward in sort of

 

  7   resolving some of those issues which have lingered

 

  8   on, and sort of adopting it as soon as possible.

 

  9             But the challenges are not trivial, and I

 

 10   tried to sort of summarize those challenges to you

 

 11   in the memorandum along with the paper that we

 

 12   wrote.

 

 13             We felt that in order to move this process

 

 14   faster and move it forward more quickly, the

 

 15   proposal to you is that we will form a working

 

 16   group under this advisory committee.

 

 17             This working group will report to you with

 

 18   their findings and provide a way forward to

 

 19   resolving the issues that have lingered on for

 

 20   three years, and come up with a very well

 

 21   structured process to resolve in a timely fashion.

 

 22             So, the proposal is a very straightforward

 

 23   proposal that this working group will report to

 

 24   you, and you will define the goals and objectives

 

 25   for this group, and you will define also the

 

                                                                46

 

  1   timeline for this group, and the proposal will be

 

  2   presented by Bob O'Neill, who is going to head for

 

  3   FDA working group members.

 

  4             Bob.

 

  5           Moving Forward -- An Approach for Resolution

 

  6             DR. O'NEILL:  Good morning.

 

  7             [Slide.]

 

  8             My name is Bob O'Neill, and as I indicated

 

  9   earlier, I am the Director of the Office of

 

 10   Biostatistics, and Ajaz and Helen have asked me to

 

 11   chair this group, which Ajaz has indicated is going

 

 12   to be reporting to you all.

 

 13             This is the process for coming to

 

 14   resolution on what you know to be a discussion that

 

 15   has been going on at least for three years under

 

 16   the specifications for delivered dose uniformity

 

 17   for inhaled and nasal drug products.

 

 18             [Slide.]

 

 19             I am going to be proposing how we are

 

 20   going to be going about doing this and asking for

 

 21   your advice and concurrence, so we can move forward

 

 22   on this.

 

 23             So, what we have thought about, and we

 

 24   have met several times with the IPAC-RS group, and

 

 25   this is the proposal.  We will have a joint working

 

                                                                47

 

  1   group under this particular committee, and it will

 

  2   be populated by senior representatives from FDA and

 

  3   from the Oral and Inhaled Nasal Drug Product

 

  4   industry, and that is mainly the IPAC group that we

 

  5   have been working with.

 

  6             [Slide.]

 

  7             The folks from FDA, I will get into the

 

  8   names in a moment, but essentially are representing

 

  9   sort of the clinical risk side of the house, the

 

 10   statistical side of the house, the generic drug

 

 11   side of the house, and the Office of New Drug

 

 12   Chemistry side of the house, so all the major

 

 13   players in terms of how this particular solution

 

 14   impacts the way we go about doing business.

 

 15             This particular proposal is essentially a

 

 16   way forward, so that we have a defined process with

 

 17   identified objectives, with identified ways of how

 

 18   we are going to communicate with each other, in

 

 19   terms of the mechanism, some timelines, some

 

 20   milestones, and how are we going to get some

 

 21   resolution on some of the issues that might be sort

 

 22   of sticky or still needing further discussion.

 

 23             So, the overall working group objective is

 

 24   to agree on a mutually acceptable parametric

 

 25   tolerance interval test for delivered dose

 

                                                                48

 

  1   uniformity, and these are the folks, and if they

 

  2   are in the room, I would ask them to stand up.

 

  3             On the lefthand side are the FDA folks.

 

  4   It is myself, Dr. Chowdhury, I believe Badrul is

 

  5   here.  He is the Pulmonary Division Director.

 

  6   Moheb Nasr, I believe is out of the country, you

 

  7   probably know him.  And Lawrence Yu, I don't know

 

  8   if Lawrence is here--there he is, and he is the

 

  9   Director for Science in Office of Generic Drugs.

 

 10             On the industry side, I think Michael is

 

 11   here, Michael Golden from GlaxoSmithKline.  Kristi

 

 12   Griffiths, I don't know if she is here, from Eli

 

 13   Lilly.  Bo Olsson from  AstraZeneca.  Dar Rosario

 

 14   from Aradigm.  Dennis Sandell from AstraZeneca

 

 15   also.  We have met with these folks and we plan on

 

 16   meeting in the future, and I will go through the

 

 17   timeline.

 

 18             [Slide.]

 

 19             So, just to reiterate, the objective of

 

 20   this working group is to develop a mutually

 

 21   acceptable, standard DDU specification, both the

 

 22   test and the acceptance criteria, for these

 

 23   products with a proposal to come back to you folks

 

 24   by the end of this year, by the end of 2004.

 

 25             [Slide.]

 

                                                                49

 

  1             So, the process that we are going to

 

  2   follow is pretty much trying to get the

 

  3   communication and the coordination of this effort,

 

  4   which is not going to be trivial, straight among

 

  5   all of us.

 

  6             We have identified that we will have a

 

  7   project manager that will help us as a working

 

  8   group stick to agendas, minutes, meeting materials.

 

  9   We plan on having monthly meetings at FDA beginning

 

 10   in May.

 

 11             The first one is probably in a few weeks,

 

 12   and in May, what we will plan to do is to review

 

 13   the feedback that you all give us today in terms of

 

 14   your blessing and what other suggestions you might

 

 15   have for how we would fine-tune this particular

 

 16   process.

 

 17             We are going to need to rely on working

 

 18   groups within the industry and within the FDA to

 

 19   further deal with the statistical issues here, the

 

 20   clinical issues, the CMC issues, and whatever else

 

 21   is on the plate, so there is likely to be some

 

 22   technical projects that will be assigned to folks,

 

 23   and the leadership and the project management of

 

 24   those particular projects will be overseen by the

 

 25   folks on the working group.

 

                                                                50

 

  1             [Slide.]

 

  2             So, again, just to reiterate the timelines

 

  3   and  the milestones, we expect to have a status

 

  4   report back to  you folks in the fall, in the

 

  5   meeting in the fall, in October, and hopefully to

 

  6   submit recommendations to you by the end of 2004

 

  7   that you can act on and come back to us on.

 

  8             [Slide.]

 

  9             Here is where we think we are to date.  We

 

 10   have discussed these issues at length and here is

 

 11   what we think we have reached consensus on.

 

 12             That the parametric tolerance interval

 

 13   approach is an improvement on the current test.  It

 

 14   is a concept that requires refinement and further

 

 15   development to address the regulatory requirements.

 

 16   There are still things that need to be fine tuned.

 

 17             We believe that there has been a lot of

 

 18   work, productive work, a lot of understanding, but

 

 19   it is time to move forward and come to closure

 

 20   particularly on this particular test.

 

 21             So the working group is formed to devote

 

 22   the necessary time and the resources to get this

 

 23   thing done, and that is through review of

 

 24   additional data analyses, especially some of the

 

 25   appropriate statistical procedures.

 

                                                                51

 

  1             [Slide.]

 

  2             We also recognize that there is some stuff

 

  3   hanging out there that needs consensus.  You have

 

  4   probably seen a presentation and heard about a

 

  5   presentation with regard to the different operating

 

  6   characteristic curves, the parametric tolerance

 

  7   interval test versus sort of the zero tolerance

 

  8   test, and there is a gap that essentially is the

 

  9   difference between the producer and the consumer

 

 10   risk, and it sort of differs in the middle over

 

 11   what you might assume to be the standard deviation

 

 12   of some of the measurements.

 

 13             That is essentially where a lot of the

 

 14   discussion has been.  Much of the discussion has

 

 15   been around what the performance characteristics

 

 16   are of the different tests under assumed scenarios.

 

 17   Another way of saying assumed scenarios is the

 

 18   simulated data, so if this, then that.

 

 19             So, if the data were to perform this way

 

 20   or lay itself out this way, then, this is what the

 

 21   operating characteristics of that particular test

 

 22   procedure are.

 

 23             So, we are actually also interested in

 

 24   seeing what real data is, so there is a number of

 

 25   issues with regard to actual data that is not in

 

                                                                52

 

  1   our hands, not in FDA's hands, which would lead us

 

  2   to say, well, how many situations are there where

 

  3   the standard deviations start to push out to 12,

 

  4   13, 14, 15, because those are the areas where you

 

  5   may be wanting to have a little more information

 

  6   because if you are not in the symmetric situation,

 

  7   your outliers are going to be where your problem

 

  8   cases are.

 

  9             So, there is some more work to be done in

 

 10   this area, so talking about that and marrying both

 

 11   the zero tolerance interval concept with the

 

 12   parametric tolerance interval idea is essentially

 

 13   where the statistical details of the test are

 

 14   likely to be focused over the next few months.

 

 15             Obviously, this issue of the applicability

 

 16   to non-normal distributions, asymmetric bimodal

 

 17   distributions, which essentially may be very much

 

 18   characteristic of manufacturing processes of, you

 

 19   know, large and small particles, and things like

 

 20   this, which is not an unusual statistical scenario

 

 21   when you have mixtures of populations, so that is

 

 22   from the statistical perspective.

 

 23             [Slide.]

 

 24             The next steps are to ask you folks to

 

 25   endorse this idea or to suggest some refinements to

 

                                                                53

 

  1   it.  We will come back to you with a status report

 

  2   as to where we are in October, and the working

 

  3   group is planning to submit recommendations to you

 

  4   all by the end of this calendar year.

 

  5             With that, I think I am done.  I would be

 

  6   willing to take any questions, and I think anybody

 

  7   on the working group would also be willing to chime

 

  8   in.

 

  9             Committee Discussion and Recommendations

 

 10             DR. KIBBE:  We have time now for

 

 11   questions, it's on our schedule, so ask questions.

 

 12             DR. SINGPURWALLA:  Well, just out of

 

 13   curiosity, what is the DDU test?

 

 14             DR. O'NEILL:  Delivered dose uniformity

 

 15   test.  It is essentially a measurement of, it's

 

 16   content uniformity, how much of the dose is

 

 17   delivered in, let's say, a spray or these nasally

 

 18   inhaled products, so it's a matter of if this was a

 

 19   pill, you would be crunching it up, you would be

 

 20   looking at what its content is, you would have a

 

 21   measure of that, and the test is essentially that

 

 22   you agree what the goalposts are for an acceptable

 

 23   amount of variability for the active ingredient,

 

 24   and if it's in that zone, it's acceptable; if it's

 

 25   not in that zone, it is not acceptable, so it's a

 

                                                                54

 

  1   variant.

 

  2             That is the whole concept behind the

 

  3   delivered dose uniformity, that the product has to

 

  4   have some consistent uniform characteristics to it.

 

  5             DR. SINGPURWALLA:  So, how would it differ

 

  6   from the parametric tolerance interval?

 

  7             DR. O'NEILL:  Well, first of all, it

 

  8   differs in a number of ways.  I don't want to go

 

  9   through the test, that there has been a

 

 10   presentation on this, and there is a lot of

 

 11   background stuff on this.

 

 12             The key difference between the zero

 

 13   tolerance is it's a zero/1 kind of thing, it's

 

 14   either in or out, and it doesn't take the standard

 

 15   deviation into account.

 

 16             The parametric tolerance interval approach

 

 17   is probably, assuming that you have something close

 

 18   to normality, and it is essentially basing the test

 

 19   both on the estimate of the mean and the estimate

 

 20   of the standard deviation, and then depending upon

 

 21   the combination of both of those guys, it is

 

 22   essentially a zone of equivalence, but the

 

 23   distinction between the two tests is one sort of a

 

 24   zero/1, you are either all in or all out, but it

 

 25   doesn't estimate the standard deviation.

 

                                                                55

 

  1             The work that has been done on the

 

  2   parametric tolerance interval approach

 

  3   statistically is intended to be a more powerful,

 

  4   more precise, take more of the information into

 

  5   account.

 

  6             DR. SINGPURWALLA:  So, would you say that

 

  7   the DDU test is not a statistical test, it has no

 

  8   statistical basis?

 

  9             DR. O'NEILL:  No, I would not say that at

 

 10   all.  In fact, both of them have statistical bases.

 

 11   In fact, the zero tolerance test is essentially the

 

 12   USP test that is used for all content uniformity,

 

 13   it is a variation on that.

 

 14             Take 10, see whether they are in the

 

 15   limits or out of the limits, if not, take another

 

 16   20.  If they are in the limits or out of the

 

 17   limits, and you are done, up or down.  That is what

 

 18   the test has been for years.

 

 19              What this is, is essentially to say,

 

 20   well, I am not using all the information, I am not

 

 21   finding out actually what the variability of the

 

 22   process is, so I want to get some handle on what

 

 23   the standard deviation of the process is, so I want

 

 24   to estimate that also, and I also want to estimate

 

 25   what the mean is.

 

                                                                56

 

  1             So, if you were to back up and sort of

 

  2   look at this within the mainstream of process

 

  3   control, you sort of want to look at where you are

 

  4   in the standard deviation world, where you are in

 

  5   the mean target close to what the center of the

 

  6   distribution is.

 

  7             So, both of these are statistical in the

 

  8   sense that they have probabilities of consumer risk

 

  9   and regulatory risk, but it is that part of it that

 

 10   is the statistical aspect of it.

 

 11             DR. SINGPURWALLA:  So, if I were to

 

 12   understand what you are saying, the DDU test seems

 

 13   like a binary test, it's a sequential binary

 

 14   process.

 

 15             DR. O'NEILL:  Well, what we are talking

 

 16   about, we are talking about the parametric

 

 17   tolerance interval test versus what is--I don't

 

 18   know what its best name is--but it would be like

 

 19   the zero tolerance interval test.  That test is

 

 20   binary.  The other one is--

 

 21             DR. SINGPURWALLA:  Is not binary.

 

 22             DR. O'NEILL:  --is not binary.  It takes

 

 23   more of the information into account.  That is the

 

 24   conceptual idea.

 

 25             DR. KAROL:  Could you tell me how much

 

                                                                57

 

  1   real data you have and what is the source of the

 

  2   real data?

 

  3             DR. O'NEILL:  Well, we have, our folks, I

 

  4   know that there are folks maybe in the audience who

 

  5   have looked at data that we have from the industry,

 

  6   but it is not necessarily the data that is all the

 

  7   data.

 

  8             I mean what we have is data that is

 

  9   submitted to us in applications and in annual

 

 10   reports, and often that is data that has already

 

 11   been screened in the sense that it either passes or

 

 12   doesn't pass, so in some sense, we are seeing data

 

 13   that is less variable than the data that these

 

 14   tests are intended to apply to uniformly.

 

 15             I believe that is where our comfort level

 

 16   is in terms of trying to understand how much

 

 17   variability is in the data, and I think it's a

 

 18   conceptual thing getting back to the way Helen

 

 19   talked about.

 

 20             For years, for years, I think the process

 

 21   was let's set the goalposts and then see whether we

 

 22   can manufacture it to fit the goalposts as opposed

 

 23   to the other way around, sort of saying what is the

 

 24   process capability and then fix the goalposts for

 

 25   the process capability.

 

                                                                58

 

  1             Under continued process improvement, the

 

  2   idea is to be closer to the target mean and to be

 

  3   closer and tighten down your variability.  It may

 

  4   be if you can't do any better, that's what you are

 

  5   left with.

 

  6             So, our situation is understanding that,

 

  7   and what we are seeing now is I believe, if I am

 

  8   not speaking for our chemists, our folks are seeing

 

  9   relatively tight standard deviations in the 5, 6, 7

 

 10   area, and the idea that there could be some

 

 11   standard deviations that are hanging out in the 12,

 

 12   13, 14 area is how come.  We are not necessarily

 

 13   seeing all of that.

 

 14             So, we want to see a little more data

 

 15   along those lines.  So, that is sort of

 

 16   conceptually where the gap is in terms of trying to

 

 17   move transitionally from the current test into a

 

 18   test that we believe has a lot more merit for

 

 19   several reasons.

 

 20             One, it captures better a handle on the

 

 21   variability of the data, and, secondly, you should

 

 22   be rewarded for taking more samples than less

 

 23   samples.  So, this test needs to reward you for

 

 24   having better estimates of what your variability is

 

 25   rather than less.  That is another conceptual part

 

                                                                59

 

  1   of this.

 

  2             DR. MEYER:  I might be mistaken because I

 

  3   don't normally read the USP, but it seems from my

 

  4   recollection there are some tablet products that

 

  5   have a specification for variability, as well,

 

  6   warfarin being an example, where you do 10, then

 

  7   you do 20, but you also look at standard deviation

 

  8   or coefficient of variation as some marker for

 

  9   approval or not.

 

 10             Is that correct?

 

 11             DR. O'NEILL:  Ajaz.

 

 12             DR. HUSSAIN:  Right, I think, Marv, you

 

 13   are right, in the sense the traditional approach,

 

 14   in the pharmacopeial approach, which are market

 

 15   standards, and they were never intended to be

 

 16   release standards, and that is the purpose they

 

 17   serve, are to maintain the market standard.

 

 18             In the case of tablets and solid dosage

 

 19   forms, you have a non-parametric approach to that,

 

 20   and, say, you have your goalposts 85 to 115 for 10

 

 21   tablets, and if one is outside that, you go to 75

 

 22   to 125 with 20 additional ones.

 

 23             For those, you have an estimate of

 

 24   standard deviation.  I think it's 6.6 person at the

 

 25   second stage, so  you have to meet that.

 

                                                                60

 

  1             The test we have for dose content

 

  2   uniformity or delivered dose uniformity for

 

  3   inhalation products right now, the FDA guidance

 

  4   doesn't have a value of standard deviations.  It

 

  5   simply says take, if it's 85 to 115, if one is

 

  6   outside that, take 20 more, and they all have to be

 

  7   within 75 to 125.

 

  8             So, the term "zero tolerance" actually is

 

  9   not really a meaningful term, and I think we

 

 10   discussed that at the previous committee, but if

 

 11   you really look at it, Jurgen had one set of

 

 12   comments at the end of that meeting, and our

 

 13   statisticians there had a very different set of

 

 14   comments on that, so we were very divided on that,

 

 15   because zero tolerance is for that sample, and that

 

 16   is, in my opinion, a big hindrance to continuous

 

 17   improvement because it forces industry to do only

 

 18   30 tests.

 

 19             If they do more, they are at risk, so that

 

 20   is not conducive to PAT, that is not conducive to

 

 21   the 21st century process that we want to move

 

 22   forward, so this actually is a model or the

 

 23   framework for what we would like to do for all

 

 24   specification, because clearly, the compendia,

 

 25   there is no movement.

 

                                                                61

 

  1             I don't see much movement in the compendia

 

  2   to change that, so we will have to move forward and

 

  3   change that, because if the compendia don't change

 

  4   that, they are going to be hindrance to PAT and

 

  5   everything else that follows.

 

  6             DR. BOEHLERT:  Just as a follow-up to

 

  7   that, I believe under ICH, the compendia are

 

  8   looking at harmonizing general chapters, and one of

 

  9   the ones they are looking at is content uniformity

 

 10   and should there be a tie-in somewhere with that

 

 11   group and what they are looking at and what they

 

 12   are doing, so you don't go two separate ways in two

 

 13   separate directions.

 

 14             DR. HUSSAIN:  I agree, but compendia are

 

 15   still a market standard, they are not a release

 

 16   standard, so from a regulatory perspective, that

 

 17   has always been the case.

 

 18             DR. BOEHLERT:  That has always been the

 

 19   case.

 

 20             DR. KIBBE:  Tom.

 

 21             DR. LAYLOFF:  I was going to say also

 

 22   there is a market standard in the way--you end up

 

 23   in a contradiction if you test the whole lot, it

 

 24   will always fail, because of the standard

 

 25   deviation, so you can't really do that.

 

                                                                62

 

  1             But in the regulatory laboratory, what we

 

  2   used to do is if we found one out of limits, then,

 

  3   we would submit it for check analysis, and if it

 

  4   passed check analysis, then, it was okay.  So, you

 

  5   sort of got around that contradiction in the limit

 

  6   setting.

 

  7             DR. KIBBE:  Anybody else?

 

  8             Is there anyone on the committee who

 

  9   thinks that moving forward is not necessarily the

 

 10   way to go?  Is there something that we need to

 

 11   discuss, because they are essentially asking us to

 

 12   say, well, yeah, we need to move forward and let's

 

 13   get the results by the end of the year?

 

 14             DR. SINGPURWALLA:  Do we have to do this

 

 15   right now?

 

 16             DR. KIBBE:  We are not going to decide on

 

 17   which tests to do right now.  We are just

 

 18   supporting the concept of having the working group

 

 19   move forward and give us a report.

 

 20             DR. SINGPURWALLA:  But one of the things

 

 21   they wanted is recommendations .

 

 22             DR. O'NEILL:  No, I don't think so.  We

 

 23   are just asking you to endorse the idea of moving

 

 24   forward and having this group, and we will come

 

 25   back to you with a report.  If  you don't like it,

 

                                                                63

 

  1   you can say go do more.

 

  2             DR. SINGPURWALLA:   I am sorry, you said

 

  3   suggest refinements in your talk, I made a note of

 

  4   it, so do you want the refinements now or later on?

 

  5             DR. O'NEILL:  No, we don't.

 

  6             DR. SINGPURWALLA:  So, you don't want

 

  7   refinements.

 

  8             DR. O'NEILL:  No, it's very high level,

 

  9   not detail oriented feedback that we would like

 

 10   from you right now.

 

 11             DR. SINGPURWALLA:  Because I would like to

 

 12   suggest refinements, but not at this minute.

 

 13             DR. O'NEILL:  I am sure we would be very

 

 14   interested in your refinements, and, in fact, I

 

 15   would certainly be interested in speaking with you

 

 16   outside of the meeting in terms of getting some

 

 17   additional ideas on this particular test, because

 

 18   again, this is a working group that is under the

 

 19   umbrella of this committee and essentially is

 

 20   coming back to the committee on behalf of the

 

 21   committee saying what do you think, because the

 

 22   committee is the one who is going to give the

 

 23   recommendations to the Agency.

 

 24             So, if you don't like the recommendations,

 

 25   then, it is totally within the committee's

 

                                                                64

 

  1   responsibilities and rights to say, you know, that

 

  2   is not what we had in mind, or that's not what we

 

  3   think is right.

 

  4             DR. KIBBE:  Let me get at some of this a

 

  5   little bit.  We have, I think, a tentative schedule

 

  6   to meet in October, and for you, the working group,

 

  7   to have your best shot prepared for us to look at

 

  8   and give you feedback on, right?

 

  9             DR. O'NEILL:  Yes, and it's not that we

 

 10   haven't thought this isn't ambitious either, but

 

 11   that's what we are   trying to work on.

 

 12             DR. KIBBE:  Is it reasonable for a member

 

 13   of this committee to forward suggestions to you in

 

 14   the interim and then have you incorporate them in

 

 15   the working group?  If you have some things that

 

 16   you would like to think through and then--

 

 17             DR. SINGPURWALLA:  Honestly, I was

 

 18   intrigued by the comment made that we invite

 

 19   suggested refinements, and for me to suggest

 

 20   refinements, I need to have a better appreciation

 

 21   for exactly what is going on.

 

 22             DR. O'NEILL:  I hear what you are saying.

 

 23   I guess maybe that was meant in terms of

 

 24   refinements to the process. Part of this is the

 

 25   process, and part of this is the content that the

 

                                                                65

 

  1   working group will be dealing with, and the working

 

  2   group already has essentially a proposal that they

 

  3   have been reacting to from IPAC-RS that has been in

 

  4   the works for a number of years, and it is that

 

  5   that is trying to be refined, those ideas are

 

  6   trying to be refined in the context of how do we

 

  7   understand what is currently sort of the operating

 

  8   characteristic curve of the current way we do

 

  9   things versus a new proposed way of doing things,

 

 10   and are they achieving where we want to be as a

 

 11   committee.

 

 12             I think that is the sense of the

 

 13   refinements.

 

 14             DR. SINGPURWALLA:  So, if the endorsement

 

 15   that you seek is for the process, and not for the

 

 16   inner workings of the process, I have no comments,

 

 17   go ahead, but if it is for the workings, then, I

 

 18   would like to think about it.

 

 19             DR. KIBBE:  I believe we are looking for

 

 20   moving ahead on the process right now.

 

 21             DR. O'NEILL:  That is what we are seeking

 

 22   from you, yes.

 

 23             DR. KIBBE:  What I hear my colleague

 

 24   saying is that he would like to have some input on

 

 25   the actual workings of the committee, with the

 

                                                                66

 

  1   thought process of the committee, and that if we

 

  2   could find some way to do that, to accommodate that

 

  3   situation within the budget constraints of the FDA,

 

  4   it would be useful.

 

  5             It always is good for a subcommittee or a

 

  6   working group of ours to have somebody from here to

 

  7   carry water for us.  You might get yourself into

 

  8   more work than you thought you were going to get

 

  9   into.

 

 10             Anybody else?  Jurgen.

 

 11             DR. VENITZ:  I am obviously in favor of

 

 12   moving forward, but I would like to give maybe

 

 13   somewhat of an unwanted recommendation, not

 

 14   necessarily a refinement.

 

 15             That is, when I look at the objectives of

 

 16   the working group, they are basically, primarily

 

 17   looking at the statistical properties of the test.

 

 18             I am recommending the group for having

 

 19   information on it, and I would encourage the

 

 20   committee to also, the subgroup, I guess, the

 

 21   working group, to also look at the clinical

 

 22   significance, in other words, in my mind, we talked

 

 23   about that last time, the clinical use is part of

 

 24   what risk-based manufacturing is all about.

 

 25             So, for example, it may be very different

 

                                                                67

 

  1   whether you are comparing inhaled insulin release

 

  2   to inhaled topical steroids, and I would like for

 

  3   that to be discussed as part of the working group.

 

  4             DR. O'NEILL:  I hear you.  Maybe I went

 

  5   through this a little too fast.  If you look at the

 

  6   constitution of the working group, Dr. Chowdhury is

 

  7   our clinical input on that, so that has been

 

  8   recognized, and that is why he is on the working

 

  9   group, to essentially put, as an overlay, the

 

 10   clinical risk structure on this, recognizing very

 

 11   much it might be product-specific, so that is his

 

 12   role.

 

 13             Lawrence Yu's role is also looking at this

 

 14   from, let's say, the generic drug implication, so I

 

 15   think the working group has been put together

 

 16   primarily to be relatively broad-minded.

 

 17             The statistical component of this is only

 

 18   one of multiple dimensions to this, but it is

 

 19   critical to understanding where we are in terms of

 

 20   the only thing that is not moving right now, which

 

 21   is the test that is on the table.

 

 22             DR. KIBBE:  Pat, go ahead.

 

 23             DR. DeLUCA:  Since this committee is going

 

 24   to be reporting back to this group, I am just

 

 25   wondering why a member of this group wasn't put on

 

                                                                68

 

  1   that committee, and it sounds like Nozer could have

 

  2   some real input into it, as well as being a link to

 

  3   this committee.  There may be some reason why you

 

  4   didn't do that, but I would certainly consider

 

  5   that.

 

  6             MS. WINKLE:  It certainly is an option.

 

  7   The way that this group is set up is basically a

 

  8   fact-finding group for the advisory committee, to

 

  9   give them the facts and the information that they

 

 10   will need to help make a recommendation on this

 

 11   test and how we want to move forward with it, but I

 

 12   think that it would be very helpful to have some

 

 13   input from Nozer.

 

 14             I think that he has some knowledge and

 

 15   some understanding and there is nothing that

 

 16   prohibits us from doing that, but we tried to set

 

 17   it up as an independent fact-finding group for the

 

 18   advisory committee.

 

 19             DR. SINGPURWALLA:  By the way, I just want

 

 20   to clarify that I didn't raise the question to

 

 21   thrust myself into this arena.  I was honestly

 

 22   asking a question, and since the matter has been

 

 23   raised by my colleague on the clinician, I would

 

 24   like to suggest that a Bayesian be on this

 

 25   particular group.

 

                                                                69

 

  1             DR. O'NEILL:  We will certainly be

 

  2   listening to you.  If you want to get into that

 

  3   discussion, we could, but one of the critical

 

  4   discussions we have been having right now is

 

  5   assumptions versus data, and Bayesians are heavy on

 

  6   the assumptions, but you have to have the data to

 

  7   support the assumptions, the game we are in, in the

 

  8   regulatory game we are in, and that is why we are

 

  9   trying to sort of get some sense of what does the

 

 10   waterfront actually look like, because it is very

 

 11   important to the behavior of the characteristics of

 

 12   this test.

 

 13             DR. KIBBE:  It is always fun to have

 

 14   statisticians discussing statistics.

 

 15             Do we have any other questions?

 

 16             Seeing no one's hand or little button lit

 

 17   up, I want to thank you very much.  We are looking

 

 18   forward to a very informative and useful report in

 

 19   October.

 

 20             My schedule says that we are supposed to

 

 21   be talking until 10:15, and we could either take a

 

 22   break now or if Ajaz promises to get finished in

 

 23   time for a break, we could move forward.  What is

 

 24   everyone's pleasure?  Naturally, the Bayesian wants

 

 25   to break.

 

                                                                70

 

  1             [Laughter.]

 

  2             DR. KIBBE:  I will give you all 15 minutes

 

  3   and then we will have Dr. Hussain.

 

  4             [Break.]

 

  5             DR. KIBBE:  Why don't you go ahead and

 

  6   start, Ajaz.

 

  7         Process Analytical Technology (PAT) - Next Steps

 

  8             DR. HUSSAIN:  Thank you.

 

  9             [Slide.]

 

 10             What I would like to do today is to give

 

 11   you a brief progress report on the PAT initiative

 

 12   and have three speakers.

 

 13             [Slide.]

 

 14             I will present a brief history to recap

 

 15   how we got here, current status and next steps.

 

 16   There are three topics that we want to share with

 

 17   you, finalizing PAT guidance, training and

 

 18   certification.  Chris Watts will make that

 

 19   presentation.

 

 20             What we are doing with respect to

 

 21   standards development.  Ali Afnan will talk about

 

 22   that.

 

 23             A topic that we have discussed twice with

 

 24   you, but we thought we would sort of bring some

 

 25   closure to that, what we have done with rapid

 

                                                                71

 

  1   microbial methods and how that has been a part of

 

  2   PAT.  Bryan Riley will talk to you about that.

 

  3             What we are hoping is, we have not really

 

  4   posed any questions, this is more of a progress

 

  5   report, status report, and we are moving forward,

 

  6   but if there is anything that you think we need to

 

  7   consider, please share this with us.

 

  8             The questions you might want to consider -

 

  9   are we on track?  Are there any recommendations for

 

 10   improving how we have approached PAT and how we

 

 11   might want to approach PAT in the future?

 

 12             [Slide.]

 

 13             The aspect that I often share is I think

 

 14   the PAT thought process has been in the Agency for

 

 15   a long time, and, in particular, a focal point for

 

 16   the discussion occurred in October of 1993.  I was

 

 17   not at FDA at that time, but Tom Layloff and others

 

 18   in St. Louis had organized a Symposium on

 

 19   Pharmaceutical Process Control and Quality

 

 20   Assurance by Non-traditional Means.

 

 21             The information I have about that is a lot

 

 22   of the focus became on near IR, and a lot of the

 

 23   focus tended to be on endproduct testing although

 

 24   the title was process control, and the discussion

 

 25   that led to sort of a very negative view of near IR

 

                                                                72

 

  1   and some of this technology came from FDA saying

 

  2   this cannot be USP methods, therefore, cannot be

 

  3   regulatory methods, which is probably more blunt,

 

  4   Tom will correct me if I am wrong.

 

  5             So, I think that was really an unfortunate

 

  6   aspect because from an FDA perspective, a lot of

 

  7   progress did not occur because of that.

 

  8             Tom and I spent a lot of time together

 

  9   thinking about this, and we saw this as an

 

 10   opportunity.  It was more of a discussion between

 

 11   an analytical chemist and an industrial pharmacy

 

 12   type, so we were putting our heads together and we

 

 13   made a presentation in the year 2000, the

 

 14   Millennium Conference in San Francisco.  I will

 

 15   just share some slides on that with you.

 

 16             Another meeting which was very important

 

 17   in the evolution of this process was the new

 

 18   technology meeting of Royal Pharmaceutical Society

 

 19   entitled Process Measurement and Control.  I

 

 20   actually met Ali Afnan and many other people who

 

 21   were then associated with the PAT at that meeting.

 

 22             [Slide.]

 

 23             The aspect I think which was important is

 

 24   this was a presentation that Tom and I did together

 

 25   at FIP meeting.  Tom had left FDA and was part of

 

                                                                73

 

  1   the USP at that time.  The title was Advanced

 

  2   Quality Control of Pharmaceuticals: In-line Process

 

  3   Controls.

 

  4             If you look at the outline, what we talked

 

  5   about then was pharmaceutical product development

 

  6   and manufacture: Building Quality In, and sort of

 

  7   design and specifications, how you approach that.

 

  8             We looked at modern in-line controls,

 

  9   potential advantages over traditional controls, a

 

 10   better approach for "building quality in," and

 

 11   talked about the need for accelerating industry and

 

 12   regulatory acceptance of modern in-line controls.

 

 13   That was the thought process before we coined the

 

 14   term "PAT," and so forth.

 

 15             [Slide.]

 

 16             In many sense, if you look at the cartoon

 

 17   there, that was the art of pharmacy manufacturing

 

 18   to the science of pharmaceutical manufacturing is

 

 19   how did we do granulation endpoint.  We reach in

 

 20   the bowl, grab a handful of granules, and look how

 

 21   they crumble, and then decided the granulation

 

 22   endpoint was reached, so we wanted to move from the

 

 23   art to more of a science-based approach.

 

 24             Our part of the PAT looked something like

 

 25   this, so if you look at that other cartoon there,

 

                                                                74

 

  1   that is how we saw it in 2000, this is what PAT

 

  2   might be.

 

  3             [Slide.]

 

  4             I think one of the critical meetings that

 

  5   I attended was a far more technical conclave in

 

  6   North Carolina.  I happened to walk into that

 

  7   meeting and G.K. Raju from MIT was talking about

 

  8   it, and that was a chance meeting that really

 

  9   provided us some of the critical information

 

 10   because I think without that, Tom and I could not

 

 11   have made any points in 2001.

 

 12             What the CAMP consortium, the MIT

 

 13   consortium helped us was to really put a value to

 

 14   this thought process, and based on that, we made a

 

 15   presentation to the advisory committee, Vince Lee

 

 16   was the chair then, is to initiate public

 

 17   discussion on application of process analytical

 

 18   chemistry tools in pharmaceutical manufacturing.

 

 19             You gave us strong support to move

 

 20   forward.  You recommended that we form a PAT

 

 21   Subcommittee.  We also, at that same meeting,

 

 22   related discussion on Rapid Microbial Testing,

 

 23   however, we did not discuss this further at the

 

 24   advisory committee, we had these discussions at the

 

 25   subcommittee, and that is the reason I brought

 

                                                                75

 

  1   Bryan Riley to come back and share with you that

 

  2   discussion again.

 

  3             [Slide.]

 

  4             But at the same time, I think Helen and

 

  5   Dr. Woodcock, we were discussing this, we felt this

 

  6   was much bigger than just an OPS issue, it had to

 

  7   be an FDA issue, so we took this to the FDA Science

 

  8   Board, and Dr. Woodcock presented that as emerging

 

  9   science issues in pharmaceutical manufacturing.

 

 10             We actually invited--I am not going to go

 

 11   through all the slides, but just to sort of

 

 12   illustrate the key presentations that occurred--one

 

 13   was the opportunity for improving the efficiency

 

 14   from G.K. Raju and then Doug Bean from

 

 15   PriceWaterhouseCooper, and we had industry

 

 16   colleagues from Pfizer who really came and helped

 

 17   us, saying that Pfizer has adopted a "Don't Use"

 

 18   and "Don't Tell" approach.

 

 19             That is the industry approach is to not to

 

 20   use new science and new technology because of

 

 21   regulatory uncertainty, or if it is needed, they

 

 22   will use it, but then they will do something for

 

 23   the regulators to say here, this is what you want,

 

 24   but we will control the process this way.

 

 25             So, we felt that was undesirable from a

 

                                                                76

 

  1   public health perspective, and we wanted to move

 

  2   forward to facilitate introduction of PAT, and we

 

  3   coined the term PAT. So, we got a very strong and

 

  4   unanimous endorsement from the FDA Science Board to

 

  5   move forward.  In fact, the Science Board also said

 

  6   that they would like to talk and give seminars on

 

  7   it, but they have not, but we did give them

 

  8   updates.

 

  9             [Slide.]

 

 10             Taking the recommendations of the advisory

 

 11   committee, this committee's recommendation. we

 

 12   issued a Federal Register Notice to invite people

 

 13   to participate on a PAT Subcommittee.

 

 14             So, we got people to apply.  We selected

 

 15   those individuals and we formed a PAT Subcommittee.

 

 16   We brought it back to this advisory committee to

 

 17   see whether the charter for the subcommittee is

 

 18   acceptable.

 

 19             You gave us valuable recommendations.  We

 

 20   formed the subcommittee, and we had three meetings

 

 21   - October, June, and February.  Tom Layloff served

 

 22   as the acting chair for the subcommittee.

 

 23             [Slide.]

 

 24             The subcommittee moved so rapidly we did

 

 25   not have an opportunity to remove the word "Acting"

 

                                                                77

 

  1   from these names, so while they were acting, the

 

  2   work was done, so we never finalized their

 

  3   positions.

 

  4             Dr. Kibbe, now the current chair of this

 

  5   committee, took the responsibility for PAT

 

  6   Applications Benefits Working Group.  Judy

 

  7   Boehlert, who is the chair for Manufacturing

 

  8   Committee, took the lead for Product and Process

 

  9   Development Working Group.

 

 10             Leon Lachman focused on Validation.

 

 11             Dr. Koch, who is now on the advisory

 

 12   committee, chaired the Working Group on PAT

 

 13   Chemometrics.

 

 14             So, these working groups provided us

 

 15   information, feedback to sort of help create a

 

 16   framework to write this guidance.

 

 17             [Slide.]

 

 18             We also, in parallel, were discussing this

 

 19   further at the FDA Science Board, and the key

 

 20   aspect was the PAT initiative was just a starting

 

 21   point to what was to follow, the 21st Century

 

 22   Initiative, and so forth.

 

 23             So, we took this discussion further to the

 

 24   Science Board, and the second Science Board

 

 25   discussion was very important.  There was a topic

 

                                                                78

 

  1   that Dr. Woodcock herself discussed, and that was

 

  2   actually something similar to what we had the

 

  3   discussion on parametric tolerance interval test,

 

  4   because the current regulatory system and the

 

  5   current pharmacopeial system is such that actually

 

  6   does not promote continuous improvement, it

 

  7   actually penalizes people for doing more testing,

 

  8   and therefore it had to change.

 

  9             So, we had to bring the concept of

 

 10   research and moving away from the current mentality

 

 11   of 75 to 125 type thinking, the market standard

 

 12   type thinking, so we had to build that consensus,

 

 13   and we got strong endorsement from the FDA Science

 

 14   Board to move forward also on that aspect.

 

 15             The other presentation, which is very

 

 16   important to remember, is that of Dr. Ray Sherzer

 

 17   from GlaxoSmithKline speaking on behalf of CAMP,

 

 18   and the thing that he pointed out, that there are

 

 19   many barriers, we need a paradigm shift, and that

 

 20   paradigm shift is necessary because the barriers

 

 21   are cultural, organizational, historical.

 

 22             The challenges are not technical, the

 

 23   technical knowhow exists.  The scientists can do

 

 24   this, but the barriers are significant cultural

 

 25   barriers and organizational barriers, and we could

 

                                                                79

 

  1   relate to that, because we had the same barriers

 

  2   in-house at FDA.

 

  3             [Slide.]

 

  4             As we were building the PAT team process,

 

  5   and you will see a lot of the thought processes

 

  6   that Helen expressed in terms of the desired goal

 

  7   that OPS wants to move in, this becomes a model or

 

  8   the pilot project for a lot of the things we have

 

  9   done.

 

 10             So, we had to build a PAT team for

 

 11   reviewers and inspectors and compliance officers,

 

 12   because this was the engine for success.  We had to

 

 13   think very carefully about this because we have a

 

 14   long history of turf issues.  We don't talk to the

 

 15   field, the field doesn't talk to us type of

 

 16   mentality, or this is my issue, field keep away

 

 17   type of thing.

 

 18             [Slide.]

 

 19             So, we actually started a team building

 

 20   exercise, so starting with a definition of team, a

 

 21   team is a group of interdependent individuals with

 

 22   complementary skills who are organized and

 

 23   committed to achieving a common purpose, applying a

 

 24   common process, and sharing a common destiny.

 

 25             Now, I think we clearly have worked on No.

 

                                                                80

 

  1   1 and 2, we haven't really worked on No. 3 yet, but

 

  2   the importance of this is the quality of the

 

  3   results we expect from the regulatory assessment,

 

  4   review, or inspection really depend on the quality

 

  5   of relationship between the reviewer and

 

  6   inspectors, and the quality of the relationship

 

  7   defines quality of thinking, and the quality of

 

  8   thinking defines quality of action that leads back

 

  9   to the quality of results we expect.

 

 10             So, this is really a complex issue and

 

 11   that has to be dealt with very carefully.

 

 12             [Slide.]

 

 13             We started the PAT process with three

 

 14   organizations:  our colleagues in Office of

 

 15   Regulatory Affairs, which are the GMP inspectors,

 

 16   Center for Drugs, and Center for Veterinary

 

 17   Medicine.

 

 18             The Center for Biologics chose not to be

 

 19   part of this, and we will discuss that further this

 

 20   afternoon whether they wish to join us or not.

 

 21             So, we formed a PAT Steering Committee,

 

 22   again reflecting all the different organizations.

 

 23   We formed a PAT Review and Inspection Team, and we

 

 24   actually recruited a small group, Raj Uppoor, Chris

 

 25   Watts, Huiquan Wu, and Ali Afnan to come and join

 

                                                                81

 

  1   OPS, so we had a very successful recruitment

 

  2   process.  We actually got Ali to take half the

 

  3   salary to come to work for FDA, and he did.

 

  4             We actually put a PAT Training and

 

  5   Coordination Team, and the training was critical.

 

  6   One of the critical aspects of the PAT Subcommittee

 

  7   was developing a curriculum for training, and then

 

  8   we partnered with three schools: a School of

 

  9   Pharmacy, a School of Engineering, and a School of

 

 10   Chemistry to bring this process together, all three

 

 11   National Science Foundation Centers for Excellence,

 

 12   Center for Process Analytical Chemistry,

 

 13   Measurement Control Engineering Center at

 

 14   Tennessee, and Center for Pharmaceutical Processes

 

 15   at Purdue.

 

 16             So, we brought the groups together and the

 

 17   training occurred, but I do want to share with you

 

 18   the challenges are cultural.

 

 19             [Slide.]

 

 20             If you look at the first picture, if you

 

 21   can see, a perfect team, right, so we wanted to

 

 22   work together, so we did want to talk to each

 

 23   other, it is important, and that is the message I

 

 24   really want to hone in, because the challenges

 

 25   right now we are facing, especially in companies,

 

                                                                82

 

  1   is this challenge.

 

  2             We have been able to overcome that in a

 

  3   small way within the PAT team, but this has to

 

  4   occur broadly, as Helen pointed out, throughout the

 

  5   Agency.

 

  6             [Slide.]

 

  7             So, I think the challenges are great, and

 

  8   we have to build teams by dancing together, and we

 

  9   did dance together--that is Joe Famulare and Doug

 

 10   Ellsworth dancing, you will never seen them dance

 

 11   anywhere else--and working as a team on smaller

 

 12   projects and building a team.  You can see Chris

 

 13   Watts smiling.

 

 14             [Slide.]

 

 15             That led to a team process that paralleled

 

 16   the efforts that we put together to develop a

 

 17   guidance.  The guidance is different, it is a very

 

 18   different guidance, it is not a "how to" guidance,

 

 19   it is a guidance developed as a framework, and the

 

 20   guidance simply outlines a framework that reflects

 

 21   analytical chemistry, industrial pharmacy,

 

 22   pharmaceutical engineering principles, but in an

 

 23   integrated way.

 

 24             What it does is it changes quite a bit of

 

 25   things each discipline might think about.  The way

 

                                                                83

 

  1   I like to say that is if you change the way you

 

  2   look at a thing, the thing you are looking at

 

  3   changes, so when Tom and I were discussing, we are

 

  4   discussing as an analytical chemist and a

 

  5   industrial pharmacy type.

 

  6             When we brought engineers in, we got

 

  7   engineering aspect, so now PAT is somewhat

 

  8   different than any of the three views of that.

 

  9             DR. SINGPURWALLA:  It is called the

 

 10   Heisenberg principle.

 

 11             DR. HUSSAIN:  Yes.  So, this is a draft

 

 12   guidance which we are finalizing, and Chris will

 

 13   talk to you about that, but I do want to sort of

 

 14   share some other thoughts.

 

 15             [Slide.]

 

 16             We had very successful workshops.  The

 

 17   Arden House conferences this year and last year

 

 18   were very successful, but they were very emotional,

 

 19   especially the one last year was very emotional.

 

 20             The emotions came out first as R&D versus

 

 21   Manufacturing, because they didn't want to talk to

 

 22   each other, and then it come out between

 

 23   pharmacists and engineers, so the engineers came up

 

 24   to me saying these pharmacist types don't know what

 

 25   they are doing, but it was necessary because it

 

                                                                84

 

  1   forced soul-searching, it forced the thought

 

  2   processes that was needed, and many companies are

 

  3   going through that right now.

 

  4             So, the emotions gave into a lot of

 

  5   rational discussion at Arden House this year, IFPAC

 

  6   meeting, ISPE meeting, PDA meetings.  Now we have

 

  7   several proposals, in fact, I expect by the end of

 

  8   this summer or the end of this year, you will see

 

  9   two complete PAT lines, two different companies,

 

 10   from crystallization to endproduct, complete

 

 11   automated manufacturing, so that is how fast two

 

 12   companies have moved, and one we have approved, and

 

 13   Bryan will talk to you about that.

 

 14             The first training session is complete,

 

 15   certification process is ongoing.  We have an

 

 16   ongoing interagency agreement with National Science

 

 17   Foundation.  We would like to explore ways of

 

 18   expanding this, and one opportunity that has been

 

 19   created is a new initiative called Critical Path,

 

 20   and we will share that with you next time.

 

 21             The Critical Path Initiative focuses on

 

 22   the need for research in three areas:  to improve

 

 23   drug development itself.  One of those is

 

 24   industrialization, that is where PAT fits in, and

 

 25   we want to use that as a means to sort of highlight

 

                                                                85

 

  1   the need for public funding for research,

 

  2   especially academic research in this area, and hope

 

  3   to do so in the next several months and years.

 

  4             We had an ongoing CRADA with Pfizer on

 

  5   chemical imaging.  Things are looking good there,

 

  6   and we hope to bring some of the results back to

 

  7   you for some sharing of that with you.

 

  8             We have ongoing communication and

 

  9   cooperation with other regulatory agencies.  Now,

 

 10   our European colleagues have formed a PAT team very

 

 11   much like ours.  They are actually going to meet

 

 12   the end of this month, and they have invited us to

 

 13   participate.

 

 14             Health Canada has met with us and they are

 

 15   very eager to sort of join our training session

 

 16   next year, the next training session that we start.

 

 17             MHLW, the Japanese are looking at it very

 

 18   intently and things are happening on the

 

 19   harmonization front with our trying to harmonize.

 

 20             [Slide.]

 

 21             Now, standards development, it was very

 

 22   important that we have a venue to develop standards

 

 23   that bring in the multifaceted structure, engineers

 

 24   have to talk to pharmacists, have to talk to

 

 25   analytical chemists.

 

                                                                86

 

  1             The way we thought that will happen is

 

  2   through ASTM, because ASTM has a lot of knowhow

 

  3   already, so we formed a committee called E55,

 

  4   Pharmaceutical Applications of PAT.  Ali Afnan will

 

  5   talk to you about that.

 

  6             There is growing external collaboration

 

  7   and emerging support structure.  ISPE and PDA are

 

  8   interested in PAT and are actually developing

 

  9   programs to cover a lot of the training needs for

 

 10   the next several years, we have PAT Group in the

 

 11   AAPS, discussion group.

 

 12             We are looking at possible collaboration

 

 13   between AAPS and ISPE to bring the material science

 

 14   and the engineers together to really focus on

 

 15   processing, strong support from IFPAC and the

 

 16   formation of an association for manufacturers.  I

 

 17   think they are struggling with some identity

 

 18   crisis.  They call it IFPACma, so I suggested they

 

 19   should call it IFPATma.

 

 20             I think this association will be helpful

 

 21   because it will house all the manufacturers of the

 

 22   sensors, the software, and so forth, and give them

 

 23   a voice, a common voice to move forward.

 

 24             When you have an association especially

 

 25   with a nonprofit association, we can partner with

 

                                                                87

 

  1   them more easily. AICHE has an extensive

 

  2   discussion, and we are building on the vision 20/20

 

  3   of AICHE especially in processing to see how that

 

  4   can be leveraged.

 

  5             A growing number of academic programs that

 

  6   focus on PAT.  Several PAT companies and training

 

  7   opportunities have emerged.  Pharmacopeias are

 

  8   interested in PAT.  Hopefully, they resolve the

 

  9   acceptance criteria first.

 

 10             PAT is now a part of the 21st Century

 

 11   Initiative and FDA's Strategic Plan, so I think

 

 12   that small crystal is starting to crystallize the

 

 13   system.

 

 14             [Slide.]

 

 15             The next step is guidance finalization.

 

 16   We are moving towards a quality system for the PAT

 

 17   process.  FDA will participate in the ASTM.

 

 18             This afternoon, we will discuss

 

 19   application of PAT to the Office of Biotechnology

 

 20   Products.  I want to sort of make sure I say this

 

 21   in a way that emphasizes the structure.

 

 22             Expand the scope of the guidance to

 

 23   include Office of Biotechnology Products.  Since

 

 24   they were not part of the training and

 

 25   certification program, the guidance is not

 

                                                                88

 

  1   applicable to them.

 

  2             The guidance is a framework guidance.  It

 

  3   applies to any manufacturing, whether it's biotech,

 

  4   whether it's automobile, whether it's anything, the

 

  5   concepts apply to any manufacturing, so it will

 

  6   apply to Office of Biotechnology Products.

 

  7             The reason that office is not within the

 

  8   scope is they were not trained and certified on

 

  9   this aspect.  So, the question to you would be how

 

 10   would we develop a training program that will meet

 

 11   their needs, and as we go to the second training

 

 12   program, that will have a more biotech focus and

 

 13   then that becomes part of the PAT process.

 

 14             I will stop my presentation and invite

 

 15   Chris to continue.  I think in the next two to

 

 16   three years, we want a sunset PAT.  What I mean by

 

 17   "sunset PAT," is that becomes a regular part of our

 

 18   CMC and GMP program, so it will merge with the rest

 

 19   of the system.

 

 20             Is two to three years the right time?  I

 

 21   think we will see, but the intention is that this

 

 22   is no longer a unique program, it is part of the

 

 23   current system.

 

 24             With that, I will stop.  If you have any

 

 25   questions, I will be glad to answer, or we could

 

                                                                89

 

  1   answer after Chris and others have talked.

 

  2                     Finalizing PAT Guidance

 

  3                    Training and Certification

 

  4             DR. WATTS:  Thank you, Ajaz, and thank the

 

  5   committee for giving me just a few minutes of your

 

  6   time to go over what we have done in terms of

 

  7   training and certification and moving toward

 

  8   finalizing the draft guidance that we put out back

 

  9   in September of 03.

 

 10             [Slide.]

 

 11             I just want to take a step back really

 

 12   quickly and just summarize some of the discussions

 

 13   that took place at this committee and the PAT

 

 14   Subcommittee in terms of defining what PAT is, and

 

 15   that will really give some background on the intent

 

 16   of the training program and what the focus was for

 

 17   the training program.

 

 18             The definition that came from this and

 

 19   subsequently made its way into the guidance was PAT

 

 20   is a system for designing, analyzing, and

 

 21   controlling manufacturing through timely

 

 22   measurements of critical quality and performance

 

 23   attributes of raw and in-process materials and

 

 24   processes.

 

 25             So, it is not just focused on any one

 

                                                                90

 

  1   analytical technique, it is not focused on

 

  2   endproduct only, it is the entire manufacturing

 

  3   process.

 

  4             When you think about PAT, process

 

  5   analytical technology, that term "analytical" more

 

  6   should be thought of as analytical thinking, not

 

  7   just simply analytical chemistry, so we made a

 

  8   point of emphasizing that analytical, when you

 

  9   think about that term, you should include not only

 

 10   chemical, but also physical, microbiological,

 

 11   mathematical, and risk analysis, all those

 

 12   conducted in an integrated manner to come up with a

 

 13   framework for controlling the manufacturing

 

 14   process.

 

 15             [Slide.]

 

 16             So, with that definition, the unmistakable

 

 17   focus of PAT is to really understand the

 

 18   manufacturing process.  What we outlined was a

 

 19   process is considered well understood when, number

 

 20   one, all critical sources of variability are

 

 21   identified and explained; number two, the

 

 22   variability is managed by the process, and,

 

 23   finally, product quality attributes can be

 

 24   accurately and reliably predicted.

 

 25             So, with that focus on process

 

                                                                91

 

  1   understanding, it brings in the concept of really

 

  2   risk management, so we consider that the level of

 

  3   process understanding is inversely proportional to

 

  4   the risk of producing a poor quality product.

 

  5             So, a well understood process then offers

 

  6   less restrictive regulatory approaches to manage

 

  7   change to different approaches to validation.

 

  8             So, if you focus on process understanding,

 

  9   we can facilitate risk-managed regulatory decisions

 

 10   and innovation, not only within the Agency, but

 

 11   within the manufacturing arena and the

 

 12   pharmaceutical industry in general.

 

 13             [Slide.]

 

 14             So, having that background, I want to now

 

 15   talk about this framework that we developed for PAT

 

 16   that came out in the guidance, and it was a

 

 17   framework, as I just mentioned, for innovative

 

 18   pharmaceutical manufacturing and quality assurance.

 

 19             We really set forth some scientific

 

 20   principles, some basic principles and concepts, and

 

 21   described some PAT tools that would support

 

 22   innovation.

 

 23             In my opinion, one of the most important

 

 24   aspects was the regulatory strategy that would

 

 25   accommodate innovation, and that the primary focus

 

                                                                92

 

  1   there was on the PAT team approach again which Ajaz

 

  2   mentioned briefly, the team approach to review and

 

  3   inspection.

 

  4             Along those lines, we developed a joint

 

  5   training and certification program, so I want to

 

  6   talk to you now about that training and

 

  7   certification program.

 

  8             [Slide.]

 

  9             You have already seen a few slide from

 

 10   Ajaz on the team building aspect, really getting to

 

 11   know one another very well, and again that included

 

 12   people from the Center for Drugs, both reviewers

 

 13   and compliance officers, the field investigators

 

 14   from the Office of Regulatory Affairs, and, of

 

 15   course, the compliance officers and reviewers from

 

 16   the Center of Veterinary Medicine.

 

 17             During this training program, it was

 

 18   important that all 15 individuals who were part of

 

 19   that initial training program, we went through

 

 20   everything together, every didactic session we went

 

 21   as a team, every practicum we went as a team.

 

 22             The team building obviously, everyone was

 

 23   involved there, so there it would really break down

 

 24   the communication barriers, which is really going

 

 25   to be key to ensuring that science-based,

 

                                                                93

 

  1   risk-based or risk-managed approach to review and

 

  2   inspection.

 

  3             A brief outline of the training program

 

  4   that we had.  Two didactic sessions, both of those

 

  5   were conducted here at the FDA, and three practica,

 

  6   again, at the University of Washington, the Center

 

  7   for Process Analytical Chemistry; Purdue

 

  8   University, Center for Pharmaceutical Process

 

  9   Research, and the University of Tennessee, the

 

 10   Measurement and Control Engineering Center.

 

 11             [Slide.]

 

 12             In summary, the first didactic that we had

 

 13   was really just to provide a general overview of

 

 14   some of the pharmaceutical processes, the

 

 15   scientific basis for some of those processes, why

 

 16   they may be necessary, to really give the team a

 

 17   feel for what some of those unit operations

 

 18   specifically may be trying to do to the material

 

 19   and what are some approaches for trying to control

 

 20   that process.

 

 21             Of course, there was some extensive

 

 22   discussion on some of that process analytical

 

 23   techniques, multivariate analysis, an in-depth

 

 24   discussion on the background of where some of the

 

 25   multivariate analysis techniques came from,

 

                                                                94

 

  1   principal component analysis, partial e-squares,

 

  2   how those can be used in terms of developing a

 

  3   control system for the manufacturing processes, and

 

  4   then finally, a general introduction to true

 

  5   process control from a process control engineer.

 

  6             After that, we went to the University of

 

  7   Washington in Seattle, The Center for Process

 

  8   Analytical Chemistry, and the focus there was

 

  9   really on sensor technology and development.  I

 

 10   think CPAC did a wonderful job of tying that in,

 

 11   giving some other industrial examples, and tying

 

 12   that into how some of these sensors may be applied

 

 13   to the pharmaceutical industry.

 

 14             [Slide.]

 

 15             To maintain continuity with the practicum

 

 16   visits, we took some of those, the sensor

 

 17   technology, some of the sensors that were being

 

 18   utilized at CPAC, and put them in the use onto some

 

 19   pharmaceutical processes at Purdue University.

 

 20             There, we really focused on some of the

 

 21   experiments that we conducted were blending, for

 

 22   example, compression, granulation, traditional

 

 23   solids processes, how some techniques were emerging

 

 24   that may be able to allow us to control those

 

 25   processes on line, really understand the impact of

 

                                                                95

 

  1   those processes on the final product quality and

 

  2   how they relate, not just to consider them

 

  3   independently, but how they relate to the final

 

  4   product quality as a whole.

 

  5             After having done our experiments at the

 

  6   second practicum at Purdue, we then took some data

 

  7   on the granulation process.  Then, when we went to

 

  8   the Measurement and Control Engineering Center at

 

  9   the University of Tennessee, we actually analyzed

 

 10   that data.

 

 11             Paul Kemperlein, who is part of MCEC,

 

 12   really walked us through, you know, what are some

 

 13   of the techniques that you maybe use, what are some

 

 14   limitations of these multivariate techniques that

 

 15   you may be want to be keeping in mind when you are

 

 16   going through the review of these applications.

 

 17             [Slide.]

 

 18             Finally, the last didactic, we tried to

 

 19   tie everything together again.  We broke up into

 

 20   teams, developed some case studies, so that we

 

 21   could really apply what we had learned throughout

 

 22   the training program, and discussed those as teams,

 

 23   a true team approach, a reviewer, compliance

 

 24   officer and investigator, and really began to

 

 25   discuss what some of the relevant issues were in

 

                                                                96

 

  1   terms of managing the review and inspection

 

  2   processes.

 

  3             That really ended the initial training

 

  4   portion, but by no means did we think it is

 

  5   complete.  I think continuing education is going to

 

  6   be vital to the success of this team, which Ajaz

 

  7   mentioned is really going to drive the success of

 

  8   PAT within the Agency.

 

  9             Along those lines, we have monthly video

 

 10   conferences with the people that are here in

 

 11   Rockville and the investigators that are in the

 

 12   field, and we try to discuss some of the relevant

 

 13   issues that are coming out, for example, some

 

 14   recent publications or some inspections, review

 

 15   issues that may have surfaced, and discussed those

 

 16   as a team, not individually as reviewers or not

 

 17   inspection issues individually as inspectors, but

 

 18   as a team.

 

 19             We also have developed a seminar series to

 

 20   discuss some publications that may be relevant to

 

 21   what we are trying to do within the PAT initiative,

 

 22   and, of course, we are using the Intranet to

 

 23   communicate some of these publications and discuss

 

 24   those on line, really, an easy way of communicating

 

 25   with the entire team.

 

                                                                97

 

  1             [Slide.]

 

  2             In summary, we have, in terms of the

 

  3   training and certification, we have completed the

 

  4   initial training program.  We are now in the

 

  5   process of conducting some lessons learned in terms

 

  6   of what we have accomplished with this, maybe some

 

  7   additional aspects that need to be considered, and

 

  8   some of those will be discussed with this committee

 

  9   this afternoon in terms of expanding the scope of

 

 10   PAT to include biotech products.

 

 11             Again, continuing education and

 

 12   involvement in the next training, I think is going

 

 13   to be critical for this group, so that we maintain

 

 14   links, not only with the team that we currently

 

 15   have, but the team that we intend to build.

 

 16             We can take some of the experience of

 

 17   those reviewers and investigators who have

 

 18   processed and will be processing some applications

 

 19   and who have gone on inspections and really share

 

 20   those with the new group that is coming in and the

 

 21   group that we currently have, so that we can

 

 22   understand maybe what is the best approach for us

 

 23   to go in terms of taking a team to do an

 

 24   inspection.

 

 25             Maybe we don't need to have all three

 

                                                                98

 

  1   people, maybe one or two should be sufficient, and

 

  2   we can do discussions over the telephone or

 

  3   videoing to handle some issue.

 

  4             Of course, we have involved the entire

 

  5   team in finalizing the guidance.  In my opinion, I

 

  6   think it was very important to get a real feel for

 

  7   how the reviewers felt about the guidance, how the

 

  8   compliance officers and how the investigators felt

 

  9   about the policy that was emerging in the guidance,

 

 10   really how that framework was going to be

 

 11   implemented because they are going to be the ones

 

 12   who are really driving things.

 

 13             They are going to be the ones who are

 

 14   enforcing the policy, not really enforcing the

 

 15   policy, but making sure that the process works as

 

 16   it should, so that it is a least burdensome

 

 17   approach to the industry.

 

 18             Within the Office of Testing and Research,

 

 19   you heard Helen mention Dr. Khan is coming on

 

 20   board, I think it is going to be important to

 

 21   maintain a link to the Office of Testing and

 

 22   Research, so that we can support policy development

 

 23   and future training if we develop some in-house

 

 24   expertise and what are some critical issues that we

 

 25   may want to be able to focus on in terms of review

 

                                                                99

 

  1   and inspection and some of the technologies that

 

  2   may be developed, if we can develop some of that

 

  3   expertise in-house, we can not only bring some of

 

  4   the training in-house, but also have some consults,

 

  5   we have expertise within the Agency that we can

 

  6   consult on a given basis.

 

  7             [Slide.]

 

  8             So, building on a little bit of the

 

  9   guidance finalization, we involved the entire team

 

 10   in the development of the guidance, and, of course,

 

 11   they are going to be involved in finalizing the

 

 12   guidance.

 

 13             The guidance was issued in September of

 

 14   03, and the public comment period extended through

 

 15   November 4th, and those comments are available on

 

 16   the docket.  You can see all, I think there were

 

 17   some two dozen companies or individuals that

 

 18   submitted comments to the guidance, and we are in

 

 19   the process of going through those and discussing

 

 20   those and addressing each one of those.

 

 21             We have included the entire team and we

 

 22   have broken the teams down into reviewers again,

 

 23   compliance officers, and investigators, and have

 

 24   those address each of those and see which comments

 

 25   they may think are most relevant and convey that

 

                                                               100

 

  1   back to the policy team, so that we can move

 

  2   forward in finalizing the guidance.

 

  3             With that, I am going to conclude this

 

  4   portion right here.  Again, I think we may have

 

  5   time for some questions afterwards, and I want to

 

  6   turn it over to my colleague, Ali Afnan, who will

 

  7   discuss the standards development process for PAT.

 

  8                      Standards Development

 

  9             DR. AFNAN:  Thank you very much for giving

 

 10   me the opportunity to be here.

 

 11             [Slide.]

 

 12             I am going to be very quick.  The outline

 

 13   of the talk is why we went with ASTM, what is ASTM,

 

 14   what is the history of the committee, where are we

 

 15   going with it, and I will give you some background

 

 16   also as to how, what Chris has just said, links

 

 17   into this process.

 

 18             [Slide.]

 

 19             Having focused on the processing, going

 

 20   away from product testing, which Chris very

 

 21   beautifully put out as PAT being process

 

 22   understanding, we had to come up with new standards

 

 23   and new ways of assessing whether a process was

 

 24   right or wrong.

 

 25             If the process was working well, then, the

 

                                                               101

 

  1   product would be right, so for that reason, we

 

  2   began to look at alternatives to the current

 

  3   specifications we were working with because

 

  4   effectively, we needed standards, not

 

  5   specifications.

 

  6             We needed a process which included all the

 

  7   interested parties and allowed them to come in for

 

  8   a balanced discussion, definition of balanced

 

  9   discussion being that we would each have one vote,

 

 10   it would have a due process, and, of course, there

 

 11   was the NTTAA Act, the National Technology Transfer

 

 12   Act, which mandates federal departments and

 

 13   agencies to use voluntary consensus standards in

 

 14   place of government standards wherever possible.

 

 15             So, having looked at all of those, we

 

 16   decided to look at ASTM, which had already been in

 

 17   dialog with our other departments in the agency.

 

 18             [Slide.]

 

 19             So, ASTM, which now they call themselves

 

 20   ASTM International, is an ANSI-accredited standards

 

 21   development organization with more than 100 years

 

 22   of experience in standard development.

 

 23             They actually generate standards, best

 

 24   practices, and guides, three different things, but

 

 25   they are all done through a peer review process. 

 

                                                               102

 

  1   Their offices are in West Conshohocken, and they

 

  2   meet regularly.  There is a committee which goes

 

  3   around to various places.  This year it is in Salt

 

  4   Lake City, and next year it is somewhere in Europe.

 

  5             [Slide.]

 

  6             The history of developing the committee

 

  7   was that through the winter and spring of 2003, FDA

 

  8   met with ASTM re: development of a new committee

 

  9   for Process Analytical Technology.

 

 10             In October of 2003, there was a meeting at

 

 11   ASTM, and then in December, the first

 

 12   organizational meeting was held at which interested

 

 13   parties from academia and industry were present.

 

 14             In January, the nomination and election of

 

 15   committee officers took place.  Again, if you are

 

 16   interested in the procedures and the processes of

 

 17   elections or how ASTM functions, the best place to

 

 18   look at is ASTM.org, World Wide Web.

 

 19             In February of this year, we had the first

 

 20   meeting of ASTM E55 Committee, and the next one is

 

 21   in Salt Lake City, 18th through 20th of May.

 

 22             [Slide.]

 

 23             What is the scope of E55?  E55 pretty much

 

 24   reflects the FDA PAT draft guidance, but the scope

 

 25   of the committee is that the scope of the committee

 

                                                               103

 

  1   shall be development of standardized nomenclature

 

  2   and definitions of terms, recommended practices,

 

  3   guides, test methods, specifications, and

 

  4   performance standards for pharmaceutical

 

  5   application of process analytical technology.

 

  6             The committee will encourage research in

 

  7   this field and sponsor symposia, workshops and

 

  8   publications to facilitate the development of such

 

  9   standards.  The committee will promote liaison with

 

 10   other ASTM committees and other organizations with

 

 11   mutual interests.

 

 12             What was quite interesting was it took

 

 13   about an afternoon to come up with that, and,

 

 14   really, we thank the industry for taking a very

 

 15   active role in coming up with that scope.

 

 16             [Slide.]

 

 17             Currently, E55 has three subcommittees.

 

 18   One is E55.01, which is PAT Systems Management;

 

 19   E55.02, which is Systems Implementation and

 

 20   Practice.  The Executive Subcommittee is 90, and

 

 21   then there is a third one, which is E55.91

 

 22   Terminology.

 

 23             [Slide.]

 

 24             The Chair and the elected officers, which

 

 25   was by ballot effectively, of E55, the Chairman is

 

                                                               104

 

  1   Don Marlowe from the Office of the Commissioner.

 

  2   The Vice Chair is Ray Scherzer from GSK.  The

 

  3   Membership Secretary is James Drennen from Duquesne

 

  4   University, and the Recording Secretary is Gawayne

 

  5   Mahboubian-Jones from Optimal Industrial

 

  6   Automation, Ltd., a system integration company.

 

  7             [Slide.]

 

  8             The Subcommittee officers.  E55.01's chair

 

  9   is Ken Leiper, Vice Chair is Gerry, the Secretary

 

 10   is Chris Watts.  E55.02 Chair is Ferdinando Aspesi

 

 11   from Aventis.  The Vice Chair, from AstraZeneca, is

 

 12   Bob Chisholm.  I am the Secretary.

 

 13             E55.91, which is the Terminology

 

 14   Subcommittee, has Larry Hecker, Abbott, as Chair,

 

 15   and Jim Fox, of GSK, as its Secretary.

 

 16             There are also 8 members at large, who

 

 17   serve on the E55 Main Executive Committee, and they

 

 18   are appointed from industry and academia.

 

 19             Thank you.

 

 20                     Rapid Microbial Methods

 

 21             DR. RILEY:  What I would like to do this

 

 22   morning is give you a brief update on the status of

 

 23   rapid microbiology methods as part of the PAT

 

 24   initiative.

 

 25             [Slide.]

 

                                                               105

 

  1             As you may know, rapid microbiology

 

  2   methods were not originally part of the PAT

 

  3   initiative.  We were sort of looking at rapid micro

 

  4   methods in a parallel track with the development of

 

  5   the PAT initiative, but finally, someone recognized

 

  6   it would make sense to have rapid micro methods as

 

  7   part of PAT, so at the October 2002 PAT

 

  8   Subcommittee meeting, there was an extensive

 

  9   breakout session dealing with rapid microbiological

 

 10   methods.

 

 11             A number of speakers discussed the

 

 12   importance of rapid microbiology methods, how they

 

 13   could fit into PAT and also the best way to look at

 

 14   rapid microbiological methods for the

 

 15   pharmaceutical industry.

 

 16             [Slide.]

 

 17             From that point on, we worked to try to

 

 18   integrate rapid microbiological methods into the

 

 19   PAT initiative because PAT had sort of a headstart

 

 20   on us.  So, the first thing we did was looking at a

 

 21   training session for rapid micro.  To do that, in

 

 22   July of 2003, here in Rockville, we had a training

 

 23   session.

 

 24             We invited people from CDER, ORA, CBER,

 

 25   and CVM to attend.  As an agenda, we had an

 

                                                               106

 

  1   overview of rapid microbiological method

 

  2   technologies, a very extensive overview.  We had

 

  3   two rapid micro method vendors come in and talk

 

  4   about their products and how they can be used.

 

  5             We also had a company come in and talk

 

  6   about their experiences of validating a rapid

 

  7   microbiological method for pharmaceutical use.

 

  8             [Slide.]

 

  9             Since the team approach is very important

 

 10   for PAT, one of the things we had to do was to form

 

 11   a rapid micro method team for PAT.  That team

 

 12   consists of Bob Coleman, expert drug investigator

 

 13   from ORA; Dennis Guilfoyle, a pharmaceutical

 

 14   microbiologist from the North East Regional

 

 15   Laboratory at FDA, Brenda Uratani, a microbiologist

 

 16   from the Office of Compliance, CDER, and myself.

 

 17             [Slide.]

 

 18             As we were doing the training and setting

 

 19   up the team, we were also in contact with a large

 

 20   global pharmaceutical manufacturer who was

 

 21   interested in using a rapid microbiology method for

 

 22   their pharmaceutical manufacturing process.

 

 23             We had a number of meetings with them to

 

 24   discuss their use of these rapid micro methods, how

 

 25   they would validate them, how they would submit the

 

                                                               107

 

  1   information to the Agency, that sort of thing, and

 

  2   these meetings culminated with a formal

 

  3   presubmission meeting with the applicant in 2003,

 

  4   where they discussed what they would submit and how

 

  5   they would submit it.

 

  6             Because what they wanted to do was to use

 

  7   some different rapid micro methods for release

 

  8   testing of a variety of non-sterile drug products,

 

  9   they wanted to use these at multiple manufacturing

 

 10   sites, it was decided that a comparability protocol

 

 11   would probably be the best way for them to submit

 

 12   this information to begin with.

 

 13             A comparability protocol is simply a

 

 14   written formal experimental protocol where, in this

 

 15   case, what they are demonstrating is that their

 

 16   rapid method is equivalent to or superior to the

 

 17   traditional method they have been using, and it

 

 18   talks also about the experiments they will do and

 

 19   also the acceptance criteria that they would want

 

 20   to use to demonstrate that equivalence.

 

 21             So, what they did after this meeting was

 

 22   they submitted two comparability protocols, one for

 

 23   product release testing for several non-sterile

 

 24   drug products, and also testing for pharmaceutical

 

 25   grade waters.

 

                                                               108

 

  1             After the approval of the comparability

 

  2   protocol for product release testing, they then

 

  3   submitted a changes being affected supplement to

 

  4   implement that rapid micro method for one of their

 

  5   non-sterile drug products.

 

  6             [Slide.]

 

  7             It was decided as part of this application

 

  8   process that an inspection would be done related to

 

  9   the rapid micro method implementation, and because

 

 10   of that, the rapid micro method team had several

 

 11   meetings, one in September of 2003, where we mainly

 

 12   discussed the comparability protocols that were

 

 13   submitted by the company, and then finally, in

 

 14   early February of 2004, we talked about the actual

 

 15   inspection itself, what we would do, how we would

 

 16   do it, that sort of thing.

 

 17             The inspection took place in late February

 

 18   of 2004.  It was led by again Bob Coleman from the

 

 19   Office of Regulatory Affairs, and Bob's experience

 

 20   and his leadership in this process was very, very

 

 21   helpful to us especially on the inspection process.

 

 22   It made it go very smoothly.

 

 23             We looked at the rapid micro method

 

 24   itself, how it was validated.  We looked at just

 

 25   the general microbiological laboratory aspect of

 

                                                               109

 

  1   the pharmaceutical manufacturing facility, and also

 

  2   looked at some of the GMPs related to the

 

  3   manufacturing of the product that they would be

 

  4   using the rapid micro method test for.

 

  5             The inspection found no significant

 

  6   problems.  There was no 43 issue as a result of

 

  7   that inspection, and we thought everything went

 

  8   well both from our standpoint, as well as the

 

  9   firm's standpoint.

 

 10             [Slide.]

 

 11             What is the future of rapid microbiology

 

 12   methods in the pharmaceutical industry?  I think

 

 13   the ultimate goal, the ideal would be real-time

 

 14   testing to provide immediate feedback.  I think

 

 15   that would be very, very helpful.

 

 16              Where are we today?  The  traditional

 

 17   micro methods require several days to several weeks

 

 18   to get results.  The current available rapid micro

 

 19   methods that are available today, and can be used

 

 20   today, significantly shorten that time to result.

 

 21             It can be as little as a day or maybe a

 

 22   little bit more than a day, and some of the rapid

 

 23   methods can give you results in as little as a

 

 24   couple of hours.

 

 25             We think even though it is not real-time

 

                                                               110

 

  1   testing, it still provides much better control,

 

  2   much better understanding of the manufacturing

 

  3   process from a microbiological standpoint and

 

  4   hopefully, can help detect and enable you to

 

  5   correct a potential problem before it becomes a

 

  6   real and serious problem as far as microbiological

 

  7   quality of the drug product is concerned.

 

  8             We are hoping that our experiences that we

 

  9   have had so far with our rapid micro method

 

 10   submission and inspection and approval process will

 

 11   encourage others in industry to also use this PAT

 

 12   regulatory pathway to look at other rapid micro

 

 13   methods and use them to improve their manufacturing

 

 14   process and understanding.

 

 15             I thank you for your attention this

 

 16   morning and I guess we will take questions of any

 

 17   presentations of this session.

 

 18            Committee Discussions and Recommendations

 

 19             DR. MEYER:  One question for Ajaz and I

 

 20   guess one for Chris.

 

 21             As the U.S. develops this PAT concept and

 

 22   begins to apply it, it seems like it is better to

 

 23   harmonize as things are being developed than after

 

 24   they are set in stone.

 

 25             Is there an effort with the Japanese, the

 

                                                               111

 

  1   Europeans, the Canadians to harmonize on the front

 

  2   end?

 

  3             DR. HUSSAIN:  Yes, in terms of I think

 

  4   there is quite a significant dialog and discussion,

 

  5   and I think the framework provides a way forward

 

  6   because as a framework, it does not get in how to,

 

  7   and harmonizing how-to guidance is a difficult

 

  8   challenge, so this is the time to do this.

 

  9             That is the reason we felt ASTM also

 

 10   provides a way forward because the devices, the

 

 11   Center for Devices, for example, utilize the ASTM

 

 12   standards, and these are international standards,

 

 13   so many of the members on the ASTM committees are

 

 14   international members right now, Europe and U.S.

 

 15   right now, and we are encouraging people from Japan

 

 16   to join in.

 

 17             So, that would be a way forward, so you

 

 18   are absolutely correct.  I mean we are trying to do

 

 19   that as you move along, and the progress has been

 

 20   significant on that.  That is what I tried to say

 

 21   is we are harmonizing without trying to harmonize.

 

 22             DR. MEYER:  My question to Chris, if I

 

 23   understood you correctly, there is about a

 

 24   15-member team, a variety of disciplines, that were

 

 25   sent through this fairly intensive training

 

                                                               112

 

  1   program?

 

  2             DR. WATTS:  Correct, yes.

 

  3             DR. MEYER:  Will that be all there is, or

 

  4   how is this going to grow to be 150 people or will

 

  5   it?

 

  6             DR. WATTS:  Well, as Ajaz mentioned, I

 

  7   think within a few years, two to three years, he

 

  8   envisions it being a regular part of the operation

 

  9   within the CMC review and GMP inspection when it

 

 10   comes to this team approach to PAT.

 

 11             We have every intention of expanding the

 

 12   training program to include more members within

 

 13   CDER, the Office of Pharmaceutical Science, Office

 

 14   of New Drug Chemistry, Office of Compliance, but I

 

 15   think the immediate need may be to expand the scope

 

 16   to include the Office of Biotechnology Products,

 

 17   which will be included in the discussion this

 

 18   afternoon.

 

 19             Based on a lot of the comments that we got

 

 20   from the guidance that we issued in September,

 

 21   there were a significant number of comments

 

 22   suggesting that we do expand the scope to include

 

 23   OBP, and as far as an immediate need, I think that

 

 24   may be more urgent in terms of expanding the team

 

 25   concept.

 

                                                               113

 

  1             DR. COONEY:  Another question on the

 

  2   education side, actually, two questions.  Could you

 

  3   comment a bit on what do you see as the important

 

  4   metrics that you use in measuring the success of

 

  5   the educational program and then could you also

 

  6   elaborate a bit on what do you see as the major

 

  7   challenges in continuing to evolve and develop the

 

  8   educational program?

 

  9             DR. WATTS:  Actually, I think one of the

 

 10   most important aspects was just the team approach.

 

 11   The technical aspects will be actually rather

 

 12   simple to address when it comes to terms of getting

 

 13   some expertise either within academic environment

 

 14   or within industry that have given technical

 

 15   expertise that can convey that to the team.

 

 16             Given the team approach, rather than

 

 17   expecting one member to have all the answers, then,

 

 18   as a team, we think we can have most of the right

 

 19   questions, we can ask most of the right questions,

 

 20   just not having one person have all the right

 

 21   answers.

 

 22             As Ali has said on many occasions, the sum

 

 23   of the team is much more than the individual

 

 24   components, so it is much more than just what each

 

 25   member brings to it.

 

                                                               114

 

  1             A real metric, again, I think the team

 

  2   approach, that was one of the most important

 

  3   aspects, can they communicate as a team, can they

 

  4   really work as a team, for example, with the rapid

 

  5   micro inspection process.

 

  6             That is relatively a novel concept when it

 

  7   comes to the regulatory environment.  Typically,

 

  8   the reviewers are responsible for review only,

 

  9   inspectors are responsible for inspection only.

 

 10   There is little, if any, communication between the

 

 11   two.

 

 12             What we are really treating it as is a

 

 13   two-way street, not just reviewers participating on

 

 14   inspection, but what are some of the key aspects of

 

 15   the manufacturing process that an inspector may be

 

 16   familiar with that they can convey to other members

 

 17   of the team.

 

 18             Really, I think the communication with the

 

 19   team is one of the most important aspects, the

 

 20   technical aspects or the scientific aspects, which

 

 21   will be a little simpler to address, I think, with

 

 22   training.

 

 23             DR. COONEY:  Just one more point.  In the

 

 24   training exercises, do you present problems of

 

 25   innovation or scenarios where you would not expect

 

                                                               115

 

  1   previously people to be able to have had all the

 

  2   answers and then ask them to try and synthesize a

 

  3   strategy or an approach?

 

  4             DR. WATTS:  Actually, some of the case

 

  5   studies that we developed are along those lines

 

  6   exactly.  During the second didactic, it wasn't

 

  7   just this is what one person did. This is the

 

  8   problem, how would you as a team think about

 

  9   solving that problem, not just regulating it, the

 

 10   problem of solving it in general.

 

 11             DR. KOCH:  I think the question of

 

 12   developing metrics will become increasing important

 

 13   just in observing the first class that went

 

 14   through, the team building indeed was there.  As

 

 15   you go to 150, it is going to be more difficult to

 

 16   dance, there is going to be more variation.

 

 17             The first group was exceptional.  If every

 

 18   one of the 150 projected fits that description,

 

 19   it's a wonderful program.  I think I have to add,

 

 20   too, the team building exercise that you went

 

 21   through before the training, that was I think

 

 22   replaced by a team building that occurred, say, if

 

 23   I look at the practicum and the didactic, it was

 

 24   quite obvious that the team members were very

 

 25   conscious to make sure that everybody on the team

 

                                                               116

 

  1   understood the technology to a working level, and

 

  2   it wasn't as if two or three came away with

 

  3   understanding it and didn't bring the others up.

 

  4             It was very obvious that by the end of the

 

  5   program, they were quite excited to move ahead, and

 

  6   that is where the problem I think in the future is

 

  7   going to come, is that as you grow the number in

 

  8   the team, you have to develop more metrics to

 

  9   evaluate how well it is going.

 

 10             A small number is relatively easy, I

 

 11   think, to build the teamwork especially as it is

 

 12   getting off the ground.

 

 13             DR. KAROL:  Bryan, I would like to ask you

 

 14   a little bit about the microbial methods.  That is

 

 15   very exciting that you are moving to real-time

 

 16   detection.

 

 17             Can you tell us a little bit about the

 

 18   processes that will be involved, what you are

 

 19   thinking of, and are there particular organisms

 

 20   that will be difficult to detect? You know, where

 

 21   are you having your problems in moving in this

 

 22   direction?

 

 23             DR. RILEY:  Well, right now I think the

 

 24   methods that we are looking at are fairly simple

 

 25   and straightforward.  We are not going to do

 

                                                               117

 

  1   anything too exotic to begin with.  A lot of the

 

  2   methods, even the rapid methods are still growth

 

  3   based, they have an enrichment step, and then an

 

  4   alternate detection method to detect fairly small

 

  5   numbers of microorganisms.

 

  6             But I think as we get into some of the

 

  7   more exotic methods that don't rely on any growth

 

  8   at all, you know, cytometry, that type of thing, I

 

  9   think the issue is going to be again how do you

 

 10   measure, you know, make sure you detect everything,

 

 11   and look at how are we going to validate that, how

 

 12   are we going to make sure that that is possible.

 

 13             DR. KAROL:  I wondered if you were moving

 

 14   into DNA technology or any of the molecular biology

 

 15   techniques now.

 

 16             DR. RILEY:  It is for some of the

 

 17   identification.  What I have talked about mainly

 

 18   has been the enumeration or

 

 19   qualitative/quantitative type tests, but certainly

 

 20   for identification, yes, a lot of people are

 

 21   looking at that using nucleic acid methods,

 

 22   sequencing, PCR, that sort of thing, for detection

 

 23   or identification of organisms, and that I think is

 

 24   becoming much more common, and it is something that

 

 25   I think we are encouraging, as well.

 

                                                               118

 

  1             DR. KIBBE:  Anybody else?

 

  2             DR. HUSSAIN:  Why don't we finish with the

 

  3   committee questions before the audience?

 

  4             DR. KIBBE:  If we could hold off for a

 

  5   second and see if there is anybody else on the

 

  6   committee.

 

  7             DR. COONEY:  I have a question on the

 

  8   rapid microbial.  Do you also have an interagency

 

  9   cooperation with Homeland Security, in this area,

 

 10   as well?  There seems to be a synergy.

 

 11             DR. RILEY:  We don't really have a direct

 

 12   formal connection at this point although one of the

 

 13   team members has been involved in that, so I am

 

 14   hoping that we can work something from that to get

 

 15   more involvement in our aspect of it.  But you are

 

 16   right, it does go together, a lot of those types of

 

 17   rapid methods that they would be interested in are

 

 18   things that we could apply, as well.

 

 19             DR. KIBBE:  Anybody else on the committee?

 

 20             [No response.]

 

 21             DR. KIBBE:  If you could come to the

 

 22   microphone and identify yourself, and then let us

 

 23   know what your question is.

 

 24             DR. CHERNEY:  Hi, I am Barry Cherney of

 

 25   the FDA.

 

                                                               119

 

  1             My question was essentially the same one

 

  2   as was just asked by the committee members, I know

 

  3   the CDC and other federal agencies, DARPA, are very

 

  4   interested in the rapid microbial techniques and

 

  5   have made actually a lot of advancement in that,

 

  6   and I was also wondering what we have done to get

 

  7   involved in those type of efforts as an overall

 

  8   approach for the Federal Government.

 

  9             DR. RILEY;  I agree.  I think we are

 

 10   starting to do that.  Certainly, within FDA, we are

 

 11   looking at some of the different centers to see

 

 12   what they are doing, but you are right, other

 

 13   government agencies have done a lot of work along

 

 14   these lines, and we need to have more of a coherent

 

 15   approach or at least cooperation and information

 

 16   sharing between the different agencies and

 

 17   different groups that are doing that, and I think

 

 18   that will be very helpful for everybody.

 

 19             DR. KIBBE:  Ajaz, you had something to

 

 20   say.  You leaned forward like you were poised.

 

 21             DR. HUSSAIN:  I think what would be useful

 

 22   is if you could share some thoughts in terms of how

 

 23   do you think we have progressed so far, especially

 

 24   Tom and Judy, and folks who were on the

 

 25   subcommittee, what we could have done better or

 

                                                               120

 

  1   what we should we be looking out for in the future,

 

  2   that would be very helpful.

 

  3             Also, as part of this, I think there are

 

  4   external leverages that really have to come

 

  5   together here, not only in the international arena,

 

  6   but also in terms of academia, in terms of public

 

  7   funding for some of the research that is needed

 

  8   especially in pharmaceutical manufacturing, and so

 

  9   forth, how do you recommend we move forward in many

 

 10   of these areas.

 

 11             DR. KOCH:  I guess I would make one

 

 12   suggestion, and that is not to lose the momentum

 

 13   that started with the training of the first group,

 

 14   and I know that the second group hasn't necessarily

 

 15   been put together yet, and there is obviously good

 

 16   reasons for that, but don't lose that momentum

 

 17   because it is a growing area.

 

 18             DR. KIBBE:  We have two observers from

 

 19   industry, what does industry think?

 

 20             MR. MIGLIACCIO:  I guess I would just

 

 21   comment on the training, that I think one of the

 

 22   frustrations that FDA has is the number of

 

 23   applications and supplements that are coming in

 

 24   from industry.

 

 25             The good news is, I think Chris had a

 

                                                               121

 

  1   slide that said PAT equals process understanding,

 

  2   and we are 100 percent behind that.  What we are

 

  3   doing now is using, in the framework that the

 

  4   guidance has provided, we are using PAT for process

 

  5   understanding, and we are putting all our resources

 

  6   into that, identifying sources of variability and

 

  7   dealing with them, not necessarily moving to

 

  8   primary control of our processes.

 

  9             So, I think there is some frustration that

 

 10   they are not seeing as many supplements.  Right now

 

 11   you probably have enough people trained to deal

 

 12   with what you are getting.  I think once our

 

 13   resources can move from process understanding and

 

 14   process capability into primary control, then, you

 

 15   will start seeing more supplements coming in and

 

 16   more new drug applications coming in.

 

 17             DR. KIBBE:  Anybody else?  Comment?

 

 18             DR. BOEHLERT:  I was going to make a very

 

 19   similar comment.  You know, there was a lot of

 

 20   initial interest.  A number of large companies very

 

 21   interested in the techniques involved with PAT

 

 22   making presentations. I am wondering if that is

 

 23   starting to wane, you know, if the FDA has seen a

 

 24   steady influx of companies asking for information

 

 25   or did it start off high and then it is sort of

 

                                                               122

 

  1   drifting off.

 

  2             The other issue on the microbiology, I

 

  3   think there is probably considerable interest on

 

  4   the part of companies in that technique, but there

 

  5   is some constraints around it right now, and those

 

  6   are compendia tests that are different, and I think

 

  7   there needs to be some interaction with the

 

  8   pharmacopeia on some of these topics because there

 

  9   are different endpoints.

 

 10             Even though you can demonstrate

 

 11   equivalency, the compendia test right now doesn't

 

 12   cover the rapid micro technique.

 

 13             DR. KIBBE:  Do you have a response?

 

 14             MR. MIGLIACCIO:  Yes.  On the is the

 

 15   interest waning, absolutely not.  In fact, the good

 

 16   news is if you have seen the transcripts of any of

 

 17   the recent industry meetings and presentations over

 

 18   the last year or so, we have gone from talking

 

 19   about concepts to talking about applications, and

 

 20   there are many more applications out there right

 

 21   now of PAT where people are either solving

 

 22   20-year-old problems or looking at a new way to

 

 23   make a new product.

 

 24             So, it is moving forward.  The interest is

 

 25   increasing exponentially right now.  It is a matter

 

                                                               123

 

  1   of once someone introduces in the public an

 

  2   application, others are grabbing onto those

 

  3   applications and bringing them home, so I think it

 

  4   is increasing significantly.

 

  5             Ajaz.

 

  6             DR. HUSSAIN:  I totally agree with that,

 

  7   and I think what we have seen is I think the

 

  8   requests we get for presentations have skyrocketed,

 

  9   so we cannot handle most of it, so we are actually

 

 10   refusing--not refusing--we are trying to be very

 

 11   selective in where we speak.

 

 12             I think others have taken up the charge

 

 13   and that is wonderful, and that is the reason why

 

 14   we feel that I think we don't have to keep speaking

 

 15   all the time, and we have other champions that have

 

 16   been created, and the champions are coming from

 

 17   industry, academia, and everywhere.

 

 18             The number of questions being asked of FDA

 

 19   is increasing, and the number of proposals that

 

 20   people are coming forward with is increasing.  So,

 

 21   right now, for example, we do not have many, we

 

 22   have seven or eight proposals right now, which will

 

 23   translate into some very focused comparability

 

 24   protocols and other aspects, so at least seven or

 

 25   eight by the end of this year.

 

                                                               124

 

  1             DR. KIBBE:  Tom.

 

  2             DR. LAYLOFF:  First of all, I think that

 

  3   the number of people trained is probably more than

 

  4   appropriate for the amount of material coming in.

 

  5             I think the industry is under an

 

  6   imperative to move to just-in-time manufacturing

 

  7   because of the model that Wal-Mart has put out, of

 

  8   essentially maintaining zero inventory at their

 

  9   level, which means that the inventory control has

 

 10   to shift back to the producer, which means that

 

 11   they have to be able to bring things more to

 

 12   just-in-time, and PAT is going to be able to handle

 

 13   that or make it better anyhow, reduce the dwell

 

 14   time, which is going to be critical for maintaining

 

 15   good supply and keeping inventory costs down.

 

 16             I think the initiative has gone very well

 

 17   so far.  It has to hatch on its own case, on its

 

 18   own time, otherwise, the momentum will fall apart.

 

 19   So, I think as the industry moves, and you move

 

 20   with it, it will develop and expand.

 

 21             DR. KIBBE:  Introduce yourself.

 

 22             DR. RITCHIE:  Gary Ritchie.  I am with the

 

 23   USP and currently the liaison with the process

 

 24   analytical technology project team that was formed.

 

 25             There were some questions or issues raised

 

                                                               125

 

  1   directed to the compendia barriers, I suppose, and

 

  2   what I just wanted to do with the committee was

 

  3   just to let them know that the project team is

 

  4   addressing some of those issues, one with respect

 

  5   to rapid micro methods, a second one with respect

 

  6   to I think the content uniformity issue, and,

 

  7   third, I guess in general, other techniques that

 

  8   may be perceived currently as general chapters or

 

  9   proposed that may be barriers, and that there is a

 

 10   work group that will be looking at those areas, and

 

 11   doing what we can do to see if we can improve or

 

 12   remove those barriers.

 

 13             I just wanted to make that comment and let

 

 14   the committee know that it is being actively looked

 

 15   at.

 

 16             DR. KIBBE:  Thank you.

 

 17             Tom, did you have something else?

 

 18             DR. LAYLOFF:  This is a comment more on

 

 19   compendia issues.  The compendia or market

 

 20   standards, the part of the law, and occasionally,

 

 21   you run into unusual circumstances because of

 

 22   incorporation of standards and laws, and probably

 

 23   the most exciting ones I have ever attended was the

 

 24   protein equivalent to nitrogen and the analysis of

 

 25   grain for protein equivalents is a kilodalton

 

                                                               126

 

  1   determination is done and the nitrogen is

 

  2   determined.

 

  3             There is a number called a PETN, the

 

  4   protein equivalent to nitrogen, the little

 

  5   multiplier.  Well, it turns out the multiplier was

 

  6   wrong, and it was a decision to change the number,

 

  7   and the number was off by 2 to 3 percent, something

 

  8   like that.

 

  9             It was one of the most heated meetings I

 

 10   have ever attended because everybody said if you

 

 11   change that number by 2 or 3 percent, you change

 

 12   the value of millions of tons of grain in ships and

 

 13   barges and warehouses everywhere.

 

 14             So, legal standards, even though they may

 

 15   not be correct, cannot be changed in a very

 

 16   cavalier fashion because they involve a lot of

 

 17   work, a lot of impact, and the same is true for the

 

 18   USP, there are many methods that are obsolete, but

 

 19   if you change them immediately, all the firms that

 

 20   have worked away from using those and validated

 

 21   against them, are now in a box of having to

 

 22   revalidate all their processes against the new

 

 23   standards.

 

 24             DR. DeLUCA:  Before making my comment, I

 

 25   would just comment I wonder what was the basis for

 

                                                               127

 

  1   that value in the first place, did it have peer

 

  2   review.

 

  3             With that little comment, you know, what

 

  4   we are talking about here, manufacturing process,

 

  5   for a long time, we have tried to bring science

 

  6   into the manufacturing area, and this is certainly

 

  7   an opportunity to do that.  I mean this requires

 

  8   science.

 

  9             I think science requires scholarly work

 

 10   and publications, and it seems that what I have

 

 11   heard today, an awful lot of work has gone into the

 

 12   PAT, but I am not so sure that we have seen

 

 13   publications coming out of this work, and I think

 

 14   this has got to get into the literature.

 

 15             So, I think we need to encourage that.

 

 16   Along those lines, we are.  We recognized this I

 

 17   guess a little over a year ago that we wanted to

 

 18   have an actual theme issue devoted to this in Pharm

 

 19   Sci. Tech, and Ajaz is the editor along with Tom

 

 20   Hale of that theme issue.

 

 21             What we are trying to get publications,

 

 22   people who are actually doing research in this

 

 23   area, and it seems with all the presentations that

 

 24   have gone on, some of the conferences and whatnot,

 

 25   that we could solicit from these people, and there

 

                                                               128

 

  1   is people around this table here who probably could

 

  2   be contributors to this, certainly, we would like

 

  3   to encourage the industry to submit their work in

 

  4   this area.

 

  5             So, I think this is essential to have

 

  6   this, to get this kind of research and science into

 

  7   the literature, the rapid microbiology methods,

 

  8   these would be great publications.

 

  9             I think the important thing about it, that

 

 10   you would have some peer review of these, so you

 

 11   wouldn't maybe make some mistakes about having a

 

 12   value for the nitrogen and protein correlation if

 

 13   you had that kind of critique.

 

 14             DR. KIBBE:  Bryan, you had a comment?

 

 15             DR. RILEY:  I just wanted to respond to

 

 16   the question about USP and possibly not meeting USP

 

 17   standards if you use a rapid micro method.

 

 18             I don't think it is as big a concern as

 

 19   some people may think it might be because even

 

 20   though some of the rapid methods may use a totally

 

 21   different basis of measurement and give you a very

 

 22   different number than the traditional USP microbial

 

 23   limits test or whatever, I think that you can

 

 24   certain compare, when you are assessing the

 

 25   usability of a rapid method, you can compare it to

 

                                                               129

 

  1   the results you are getting with the USP method and

 

  2   certainly set your acceptance criteria based on the

 

  3   fact that you are looking at different numbers, and

 

  4   that even though a product can still meet your

 

  5   acceptance criteria with a rapid method, it would

 

  6   still meet the acceptance criteria if you use the

 

  7   USP method even though the numbers may be very

 

  8   different.

 

  9             So, I think that should be taken into

 

 10   account and compared when you are assessing the

 

 11   method itself.

 

 12             DR. BOEHLERT:  I agree, I think the issue

 

 13   is around equivalent to or better, which is how USP

 

 14   defines alternate tests.

 

 15             DR. RILEY:  Yes, and I think demonstrating

 

 16   equivalence to the USP test should not be that

 

 17   difficult for a lot of the rapid methods.

 

 18             DR. LAYLOFF:  With regard to the testing

 

 19   for viable organisms, the rapid tests will

 

 20   frequently give false positives.  Do they also give

 

 21   false negatives?

 

 22             DR. RILEY:  It can depend on the test and

 

 23   what you are testing.  It is something that has to

 

 24   be looked at on a case-by-case basis, if you are

 

 25   looking at a product or you are looking at water,

 

                                                               130

 

  1   you could have interference, that sort of thing.

 

  2   It really depends on what you are looking at.

 

  3             As I said, there are some growth-based

 

  4   rapid methods, and those would have very

 

  5   similar--if you are looking at growth in the media

 

  6   or not, that is going to be very similar to the

 

  7   growth-based traditional compendia test.

 

  8             Some of the rapid methods that don't

 

  9   require growth, it looks like a viable stain, that

 

 10   type of thing, that is something that we would have

 

 11   to determine experimentally.

 

 12             DR. LAYLOFF:  But that would be a false

 

 13   positive rather than a false negative, or do you

 

 14   get false negatives also?

 

 15             DR. RILEY:  I think it depends on the

 

 16   method.

 

 17             DR. SINGPURWALLA:  You wanted to answer

 

 18   two questions, are we on the right track and any

 

 19   recommendations.  Well, I just need a point of

 

 20   clarification.  It has much to do with I don't

 

 21   understand what PAT is all about.

 

 22             So, the first question to you is how is it

 

 23   different from process control practiced in

 

 24   automobile industries and manufacturing industries,

 

 25   and if it is the same, I am surprised that the drug

 

                                                               131

 

  1   industry has not been using it because my sense is

 

  2   that the drug industry has been using it ever since

 

  3   I was a student.

 

  4             MR. MIGLIACCIO:  What has happened over

 

  5   the last five, seven years is we have the

 

  6   analytical technology, so the near infrared has

 

  7   been there, and statistical process control has

 

  8   been there.

 

  9             What has been absent is the engineering

 

 10   solution to bring the technology right to the shop

 

 11   floor to marry the analytical technology to the

 

 12   manufacturing equipment.  That is what we have now

 

 13   in process analytical technology.

 

 14             So, you are doing real-time process,

 

 15   monitoring, and control versus taking samples,

 

 16   bring them through a laboratory, and then doing SPC

 

 17   on that.

 

 18             So, there is a paradigm shift that we have

 

 19   gone through, that you have real-time monitoring,

 

 20   and not just of a unit dose sample that you have

 

 21   taken out of a blender or 10 tablets that you have

 

 22   taken off a tablet press, but of a very large N.

 

 23   The N has increased substantially our ability to

 

 24   monitor the process.

 

 25             DR. HUSSAIN:  I think that is a good

 

                                                               132

 

  1   point. At the same time, I think the key aspect has

 

  2   been that in the sense some have regarded that the

 

  3   pharmaceuticals would be quite different, I mean if

 

  4   you really look at some of the literature, the

 

  5   thought process had been that pharmaceutical dosage

 

  6   forms are different from making machines, and so

 

  7   forth, so some of those principles might not apply.

 

  8             So, it has been an evolution, it has been

 

  9   a paradigm shift, and in many ways, I have used the

 

 10   phrase testing to document quality to quality by

 

 11   design.  We have always talked about quality by

 

 12   design, but our mentality has been testing to

 

 13   document quality, because that is what we could do.

 

 14             I think the pharmacopeial structure, the

 

 15   regulatory structure had sort of reinforced that

 

 16   thought process on that, and Gerry is right in

 

 17   terms of when you bring the analytical tools, the

 

 18   engineers, everybody together, it is a paradigm

 

 19   shift, and it is happening now to a large degree.

 

 20             DR. SINGPURWALLA:  So, am I correct in

 

 21   understanding that you are using what the engineers

 

 22   called "control theory" techniques into the

 

 23   pharmaceutical industry, which was not there early

 

 24   on?

 

 25             DR. HUSSAIN:  I think "not there" is not

 

                                                               133

 

  1   probably the correct characterization in the sense

 

  2   different segments have different levels of

 

  3   controls, for example, manufacture of the drug

 

  4   substance material API, which is more closer to

 

  5   chemical synthesis, chemical industry, you have a

 

  6   lot more of that in there.

 

  7             Biotechnology evolved later on, so they

 

  8   have more of that already in place, because process

 

  9   is so critical.  So, there are segments, the

 

 10   pharmaceutical dosage forms, you know, tablets,

 

 11   capsules, and so forth, have not received the same

 

 12   level of attention, and that is new for these

 

 13   dosage forms, so it depends on which part of

 

 14   industry you look at.

 

 15             DR. SINGPURWALLA:  So, to come back to

 

 16   your original thing, about your question, so when

 

 17   you say PAT, this is a generic thing.

 

 18             DR. HUSSAIN:  Yes.

 

 19             DR. SINGPURWALLA:  Not specific to the

 

 20   drug industry.

 

 21             DR. HUSSAIN:  Well, the framework is

 

 22   generic to manufacturing irrespective of which

 

 23   manufacturing.  The language, the vocabulary we

 

 24   have used in the guidance is pertaining to the

 

 25   pharmaceutical industry, and from that perspective,

 

                                                               134

 

  1   it is somewhat focused on the pharmaceutical

 

  2   situation or scenario.

 

  3             DR. KIBBE:  Anybody else?  You are doing

 

  4   so well.

 

  5             In light of the fact that we have run out

 

  6   of steam, what I propose we do is break for lunch.

 

  7   We have already checked, I hope we have checked,

 

  8   with our open hearing individuals, and we are going

 

  9   to try to start the open to the public at 12:30

 

 10   instead of at 1 o'clock, so that you are all

 

 11   invited to be back here at 12:30.

 

 12             [Whereupon, at 11:15 a.m., the proceedings

 

 13   were recessed, to be resumed at 12:30 p.m.]

 

                                                               135

 

  1             A F T E R N O O N  P R O C E E D I N G S

 

  2                                                   [12:30 p.m.]

 

  3                       Open Public Hearing

 

  4             DR. KIBBE:  We have how many people who

 

  5   have asked to speak?  We have four.  Their

 

  6   presentations, if they have slides, and what have

 

  7   you, will be on the web page by tomorrow, we hope,

 

  8   so that those of you in the public who need copies,

 

  9   and what have you, can get access that way.

 

 10             We have the Regional Director of the CMC

 

 11   Regulatory Affairs from GlaxoSmithKline, Leo

 

 12   Lucisano.  We are really lucky to have you here.

 

 13             MR. LUCISANO:  I don't have any slides

 

 14   today.  Really, my comments are intended to

 

 15   complement Dr. Riley's presentation on rapid micro

 

 16   methods this morning.

 

 17             It wasn't until I saw Dr. Winkle's metrics

 

 18   this morning that I realized that GlaxoSmithKline

 

 19   provides about 10 percent of the annual reports

 

 20   that is reviewed by new drug chemistry and about 5

 

 21   percent of the manufacturing supplements.

 

 22             So, we create a lot of work for the Office

 

 23   of Pharmaceutical Sciences.  So, I was delighted

 

 24   when, on February 27th, the PATRIOT team from FDA,

 

 25   the PAT Review and Inspection Team, completed a

 

                                                               136

 

  1   week-long inspection at our facility in Parma,

 

  2   Italy.

 

  3             It was led by Bob Coleman, as Dr. Riley

 

  4   mentioned.  Bob is a national expert based in the

 

  5   Atlanta District Office, and he was accompanied by

 

  6   three microbiologists, one of which was Dr. Riley.

 

  7             The inspection actually was triggered by

 

  8   the submission of a supplemental new drug

 

  9   application in which we sought approval of one of

 

 10   the types of applications for rapid micro methods.

 

 11   That technology was endorsed as PAT technology by

 

 12   this committee back in October of 2002.

 

 13             The inspection was a success and now it

 

 14   enables us to potentially implement this technology

 

 15   across a global supply chain, and it represents the

 

 16   culmination of a 16-month effort between the Office

 

 17   of Pharmaceutical Sciences, the Office of

 

 18   Regulatory Affairs, and industry in addressing the

 

 19   challenges associated with the first PAT

 

 20   application approved as part of FDA's Quality

 

 21   Initiative for the 21st Century.

 

 22             What I wanted to do today was just spend a

 

 23   few minutes talking about the challenges that we

 

 24   had in working with the Agency to reach this

 

 25   milestone.  They were really of three types.  There

 

                                                               137

 

  1   was the technical challenges, the regulatory

 

  2   management challenges, and the educational

 

  3   challenges.

 

  4             Just by way of background with respect to

 

  5   microbiological testing in the industry, we use it

 

  6   in a lot of different ways.  We may use it to meet

 

  7   the regulatory specifications to release a drug

 

  8   product, we may apply it to the testing of

 

  9   excipients, such as water for injection prior to

 

 10   its use in the drug product, and we may also

 

 11   utilize it to verify that a manufacturing suite is

 

 12   sufficiently clean prior to the next phase of

 

 13   production.

 

 14             So, the conventional methods typically

 

 15   take about four to seven days to complete and

 

 16   oftentimes really represents the rate-limiting step

 

 17   in our decision process associated with batch

 

 18   release or release of a manufacturing area.

 

 19             So, with the availability of commercial

 

 20   instrumentation for rapid micro methods being

 

 21   available and providing results in a matter of

 

 22   hours using technologies, such as ATP

 

 23   bioluminescence and solid phase cytometry, there

 

 24   are tremendous opportunities for us in utilizing

 

 25   rapid micro methods.

 

                                                               138

 

  1             So, the technical challenges.  One of the

 

  2   examples that we had was trying to apply rapid

 

  3   micro methods to regulatory specification for a

 

  4   class of dosage forms, so in working with Dr. Peter

 

  5   Cooney's staff in the Office of Microbiology, we

 

  6   agreed on an approach that adopted a two-stage

 

  7   acceptance criteria, combining the qualitative

 

  8   rapid micro method with the currently approved

 

  9   microbial limit test that used more conventional

 

 10   methods as stated in the USP, so if a batch of drug

 

 11   product tested free of bioburden using the

 

 12   qualitative rapid micro test, that no further

 

 13   testing was required.

 

 14             If the screen indicated the presence of

 

 15   microorganisms, then, the conventional microbial

 

 16   limit test was performed to determine compliance

 

 17   with the regulatory specifications.

 

 18             So, when you think about a high-volume

 

 19   product where the historical data indicates that

 

 20   the product typically is free of bioburden,

 

 21   applying rapid micro methods in this strategy

 

 22   offers very significant advantages with respect to

 

 23   product release and inventory turnover.

 

 24             The foundation for the validation of this

 

 25   methodology was actually provided by the PDA

 

                                                               139

 

  1   technical report published in 2002.  This document

 

  2   addressed the evaluation, validation, and

 

  3   implementation of new microbiological test methods,so,

 

  4   speaking to Dr. DeLuca's comment this morning

 

  5   about the availability of published literature

 

  6   actually facilitating working through some of the

 

  7   issues that we had around process analytical

 

  8   technology and its approval.

 

  9             The second type of challenge that we had

 

 10   was the regulatory management process.  We were

 

 11   interested in applying rapid micro methods in a

 

 12   variety of different ways at multiple FDA-approved

 

 13   facilities, so this scope of interest would

 

 14   potentially affect the entire approved product

 

 15   portfolio expanding over 140 approved new drug

 

 16   applications.

 

 17             So, in the conventional regulatory review

 

 18   and approval process, this might require an

 

 19   equivalent number of new drug applications, each

 

 20   containing a data package demonstrating the

 

 21   application of rapid micro methods to the specific

 

 22   product of interest.

 

 23             So, how would we progress rapid micro

 

 24   methods without further overburdening the Office of

 

 25   New Drug Chemistry with additional supplemental new

 

                                                               140

 

  1   drug applications?

 

  2             Actually, the solution was provided by the

 

  3   Agency with the issuance of the draft guidance on

 

  4   comparability protocols that was published in

 

  5   February of 2003.  So, a comparability protocol is

 

  6   essentially a plan that evaluates the effect of

 

  7   changes on an approved product.

 

  8             You don't have to include product-specific

 

  9   data, but describe the analytical procedures that

 

 10   you intend to use and the acceptance criteria that

 

 11   will be achieved to demonstrate that there is no

 

 12   adverse effect on product quality.

 

 13             So, what we agreed upon that we would

 

 14   submit a plan, a comparability protocol to apply

 

 15   this technology, and we do it by a prior approval

 

 16   supplement.

 

 17             Once the Agency approved that plan, we

 

 18   could then implement that technology at any GSK

 

 19   manufacturing site that had a satisfactory CGMP

 

 20   inspection status with the FDA, so that if these

 

 21   conditions were met, any site within the

 

 22   manufacturing network could adopt rapid micro

 

 23   methods according to its own timeline and notify

 

 24   the Agency via a regulatory submission that does

 

 25   not require prior approval, such as an annual

 

                                                               141

 

  1   report or changes being effected in zero day

 

  2   supplement.

 

  3             This agreement applied regardless of the

 

  4   number of NDA-approved sites or number of

 

  5   NDA-approved products and dosage forms manufactured

 

  6   at a particular facility.

 

  7             So, the process, the end result was a

 

  8   streamlined management process for adopting rapid

 

  9   micro methods or really any process analytical

 

 10   technology, for that matter, across a global supply

 

 11   chain.

 

 12             It offered advantages for the Agency by

 

 13   decreasing the number of prior approval supplements

 

 14   that needed to be reviewed, and also retained the

 

 15   appropriate checks and balances for the Agency to

 

 16   conduct an inspection at its discretion and verify

 

 17   that the manufacturing site has fulfilled the

 

 18   validation requirements approved in a comparability

 

 19   protocol.

 

 20             The last challenge was one of education,

 

 21   and as the discussions evolved between GSK and FDA,

 

 22   we recognized that there was a need for both the

 

 23   Agency and GSK to educate their personnel regarding

 

 24   rapid micro methods, their science, and their

 

 25   regulation.  This was achieved in a number of

 

                                                               142

 

  1   different ways.

 

  2             Dr. Riley mentioned this morning about a

 

  3   day-long seminar that the FDA conducted with a

 

  4   large number of FDA staff, talking about

 

  5   instrumentation, bringing in consultants,

 

  6   discussing their application.

 

  7             We also had a half-day technical meeting

 

  8   between GSK scientists and FDA scientists in May of

 

  9   last year with the objective of that meeting to

 

 10   share the ongoing work that was evolving with rapid

 

 11   micro technology, but there was also a need to

 

 12   educate the global organization within GSK since

 

 13   the regulatory process that was approved for rapid

 

 14   micro methods was very different from the

 

 15   conventional post-approval process for implementing

 

 16   changes.

 

 17             We also had to review our changed

 

 18   management systems to assure that they could

 

 19   accommodate the conditions of approval agreed upon

 

 20   with the agency.  The regulatory management process

 

 21   approved for the implementation of rapid micro

 

 22   methods has implications for other process

 

 23   analytical technologies in the future.

 

 24             Consequently, we have functional groups

 

 25   within my organization requesting the Regulatory

 

                                                               143

 

  1   Affairs Department to educate them under rapid

 

  2   micro experience with FDA, and to guide them with

 

  3   respect to future PAT applications and their

 

  4   approval.

 

  5             Sixteen months from the time that this

 

  6   advisory committee endorsed rapid micro methods as

 

  7   a process analytical technology, we now have an

 

  8   approved regulatory process that can be applied

 

  9   across the company's entire product line at any

 

 10   FDA-approved facility.

 

 11             It required the review and approval of

 

 12   three supplemental new drug applications and an FDA

 

 13   inspection by the PATRIOT team.

 

 14             I would like to thank this advisory

 

 15   committee for really providing the incentive to

 

 16   address the technical, the regulatory management

 

 17   and educational challenges associated with the

 

 18   approval and implementation of a PAT technology

 

 19   platform.

 

 20             The resolution of these challenges

 

 21   required the application of new guidance documents,

 

 22   such as the guidance on comparability protocols,

 

 23   the availability of published scientific

 

 24   literature, such as PDA Report 33, and a new way of

 

 25   conducting business that really required some

 

                                                               144

 

  1   introspection and some adjustment on both the

 

  2   Agency's part and ours.

 

  3             I know within GSK, we are now motivated by

 

  4   these first approvals and are looking at additional

 

  5   applications of process analytical technologies

 

  6   that may be more expansive in scope and require a

 

  7   somewhat different road map, but I suspect the

 

  8   challenges will still be somewhat the same and

 

  9   require a similar investment of resources, cost,

 

 10   and flexibility to be successful.

 

 11             Thank you.

 

 12             DR. KIBBE:  Do you have any questions for

 

 13   our speaker, anybody?

 

 14             [No response.]

 

 15             DR. KIBBE:  Well, I will hit you with one.

 

 16   Do you have an estimate of what was saved in

 

 17   man-hours or paperwork on both ends of the street,

 

 18   like for your people and for the FDA people?

 

 19             MR. LUCISANO:  With respect to reductions,

 

 20   at the FDA inspection at Parma, we shared with the

 

 21   Agency that for one particular product, we would be

 

 22   saving 160 kiloEuros--it was a European site--per

 

 23   year with the application of rapid micro methods.

 

 24             Certainly, the availability of only

 

 25   submitting or the opportunity to submit only two

 

                                                               145

 

  1   supplements rather than 140 supplements to gain

 

  2   approval of a technology across approved product

 

  3   line offers significant cost reductions for the

 

  4   Regulatory Affairs Department.

 

  5             DR. KIBBE:  Anybody else?

 

  6             [No response.]

 

  7             DR. KIBBE:  Thank you.

 

  8             We now have two individuals from

 

  9   Xcellerex, the founder, Parrish M. Galliher, and

 

 10   the Vice President, Quality and Regulatory Affairs,

 

 11   Elizabeth Fowler.

 

 12             MR. GALLIHER:  Good afternoon.  Thank you

 

 13   to the advisory committee and Keith Webber in

 

 14   particular for the invitation, and Ajaz's support

 

 15   and vote of confidence for our view on PAT for

 

 16   biologics.

 

 17             [Slide.]

 

 18             I want to introduce Beth Fowler, who is

 

 19   our VP of Regulatory and Quality at Xcellerex.

 

 20             The title of our talk is PAT for

 

 21   Biologics, Ensuring Quality of Biologically

 

 22   Produced Drugs.

 

 23             I think I want to focus, what I would like

 

 24   to sort of conduct as sort of a brainstorm view of

 

 25   our opinions on PAT, to focus in on biotech drugs,

 

                                                               146

 

  1   recombinant proteins produced in mammalian cell

 

  2   systems or bacterial yeast systems, and less on the

 

  3   traditional biologics, such as vaccines.

 

  4             So, before I get going into what we have

 

  5   to say, I would like to mention that PAT, to us, is

 

  6   much broader than the title, the words of PAT.  It

 

  7   is not just, in our view, process analytical

 

  8   technology that we are concerned about, it is a

 

  9   broader vision of ensuring product quality across

 

 10   all stages of manufacturing, but also throughout

 

 11   the organization that is responsible for delivering

 

 12   the drug that comes from all parts of the

 

 13   organization as the process is developed, as the

 

 14   process is implemented, and as the product quality

 

 15   is assessed either in-line, at-line, or off-line.

 

 16   We will talk a bit about how, at Xcellerex, we are

 

 17   trying to take this broader view as part of doing

 

 18   business for ourselves and for our customers.

 

 19             We are a contract manufacturing

 

 20   organization for biotech products, so we have the

 

 21   customers' product quality right square in our

 

 22   view, and that drives a lot of what we do in our

 

 23   business, and we find that PAT, in general, in the

 

 24   broader interpretation, is really good business for

 

 25   us and for our customers.

 

                                                               147

 

  1             I think looking back over the last 25

 

  2   years of the biologics manufacturing business, I

 

  3   think in the eighties, the industry was consumed

 

  4   with the simple task or the herculean task of

 

  5   simply producing these products and the initial

 

  6   ones getting to market, and I think the industry is

 

  7   really consumed with that whole endeavor, which was

 

  8   huge.

 

  9             In the nineties, more and more drugs,

 

 10   biotech drugs, came to the market.  We now have

 

 11   approximately 30 individual proteins that have been

 

 12   licensed, so getting to market in the nineties was

 

 13   really where the industry was focusing.

 

 14             However, in the last five years, we have

 

 15   seen the importance of speed getting to the clinic

 

 16   and speed getting to the market as being more and

 

 17   more of a driver in what we hear from our

 

 18   customers, what we have seen in our own lives and

 

 19   biotech companies, rushing drugs to the clinic and

 

 20   to the market, and very much our interpretation

 

 21   today of PAT is focused, not just on quality, but

 

 22   also affording speed without sacrifice of quality

 

 23   or, in fact, improving quality, and I will talk a

 

 24   bit more about that.

 

 25             However, in the next decade, we see the

 

                                                               148

 

  1   indications and trends in the industry impacting or

 

  2   bringing costs to the forefront of biotechnology

 

  3   and manufacturing through the advent of competition

 

  4   with a number of similar compounds in the market,

 

  5   through the pressures of managed health care, and

 

  6   so we think that PAT will actually be invigorated

 

  7   or stimulated by cost pressure of the industry

 

  8   coming in the next decade.

 

  9             [Slide.]

 

 10             In general, that was our review of PAT,

 

 11   again a broader vision than simply process

 

 12   analytical technology.  We are going to talk about

 

 13   the importance of PAT specifically for biologics

 

 14   manufacturing and drill down into the real value

 

 15   and how we are, in several places throughout the

 

 16   organization, in our business, implementing PAT

 

 17   principles at various levels.  I will give you

 

 18   specific examples of those, and then wrap up with

 

 19   some conclusions.

 

 20             [Slide.]

 

 21             We view PAT as process knowledge gained

 

 22   through process analytics and statistically

 

 23   designed process optimization studies to begin

 

 24   with.  So, in our view, it really begins with

 

 25   understanding the process that is intended to

 

                                                               149

 

  1   deliver a product of a certain quality.

 

  2             So, we view PAT more as product quality

 

  3   knowledge rather than process analytical

 

  4   technology.  The focus is really on product quality

 

  5   wherever it is being produced in the manufacturing

 

  6   process.

 

  7             Again, to begin with, we start in the

 

  8   development laboratory by studying the parameters

 

  9   that affect product quality and yield in a

 

 10   statistically oriented fashion in robustness

 

 11   studies, and I will get into that a little bit.

 

 12              So, the processes are really designed to

 

 13   maintain product quality or to, in fact, improve

 

 14   product quality, and we make real efforts there,

 

 15   and I will give you some examples.

 

 16             We see the advent of continual monitoring

 

 17   to, in fact, further ensure process control to

 

 18   produce a product of a defined quality, and the

 

 19   reason that we think continual monitoring is a

 

 20   benefit is, in fact, that if there are process

 

 21   perturbations during a lengthy step, we can analyze

 

 22   those perturbations more quickly and determine

 

 23   whether or not that product is in jeopardy or

 

 24   whether, in fact, we should invest in further

 

 25   processing to carry it to final form.

 

                                                               150

 

  1             With regard to then the manufacturing risk

 

  2   of further processing a batch that could be in

 

  3   danger, again, PAT, with the database that is

 

  4   generated through the efforts of PAT, will help us

 

  5   not only assess the risk to product quality, but

 

  6   also from a manufacturing economic side, is it

 

  7   worthwhile investing in a batch that has some sort

 

  8   of perturbation in this processing step.

 

  9             So, it is not just risk to product

 

 10   quality, but in point of fact, from a

 

 11   manufacturer's standpoint, we are also concerned

 

 12   about are we delivering what the customer wants,

 

 13   are we delivering on the contract to produce a

 

 14   certain amount of product of a certain defined

 

 15   quality.

 

 16             [Slide.]

 

 17             Just to talk a little bit about some of

 

 18   the key issues that we see in this business, first

 

 19   of all, just stepping back a bit, there is

 

 20   biological variation in production of the material

 

 21   that we are interested in depending on the system

 

 22   with which you are producing the product.

 

 23             If you are in a transgenic system, there

 

 24   can be animal to animal variation, and in cell

 

 25   culture based processes, whether they be mammalian,

 

                                                               151

 

  1   bacterial, or yeast or fungi, there is variation in

 

  2   the cell culture step.

 

  3             It is not a surprise, the organisms are

 

  4   very complex, they have a huge number of genes

 

  5   whose function can impact the manufacture of the

 

  6   product, so we expect that in biological systems,

 

  7   there will be inherently more variation that could

 

  8   affect product quality.

 

  9             There can also be unknown pathogens

 

 10   associated with biological systems, and these, of

 

 11   course, are an issue with regard to biological

 

 12   safety of the product, and there can be, in fact,

 

 13   unrelated impurities to the drug with unknown

 

 14   activities that can, in fact, be produced by

 

 15   biological systems at low quantities that may not

 

 16   be measured.

 

 17             So, in general, we see biologics as a

 

 18   highly variable environment within which to make a

 

 19   product, and taking this account, it is to me quite

 

 20   remarkable and wonderful that there are so many

 

 21   approved products on the market today helping so

 

 22   many people who are, in fact, in need.

 

 23             So, we see this background therefore

 

 24   advocating the greater value then of more process

 

 25   analytical technology  or more product quality

 

                                                               152

 

  1   knowledge given the inherent variability.  So, that

 

  2   is the general background in which we see the value

 

  3   of PAT.

 

  4             [Slide.]

 

  5             To just talk a bit more about product risk

 

  6   management, this is our present state of affairs.

 

  7             First of all, in today's biologics

 

  8   manufacturing, we generally start with a viewpoint

 

  9   that minimal process change should be attempted or

 

 10   even allowed as the drug moves through the clinic

 

 11   or into the clinic and on to the market, we try to

 

 12   minimize the number of process changes.

 

 13             Two.  Process parameters are used, that

 

 14   is, process control parameters are generally used

 

 15   as surrogates for product quality indication or

 

 16   monitoring.  That is, we are not really directly

 

 17   monitoring the product quality attributes in every

 

 18   step of the manufacturing process mainly due to

 

 19   limitation in analytical technology and specificity

 

 20   especially in the presence of crude background

 

 21   materials and matrices that interfere with current

 

 22   methodologies.

 

 23             Therefore, we rely on post-production

 

 24   release and some in-process testing again through

 

 25   surrogate markers to ensure product consistency. 

 

                                                               153

 

  1   Again post-facto is the point, post-production is

 

  2   the operative here.

 

  3             Generally, we are faced with processing a

 

  4   batch to completion, which can be an investment of

 

  5   millions of dollars, and then to find out that we

 

  6   have somewhere along the way lost the product

 

  7   quality attributes that we intended to achieve, and

 

  8   that batch no longer can be considered releasable.

 

  9             So, today's business is post-production

 

 10   and there is a huge investment made in the intent

 

 11   of that batch being released, but, in fact, our

 

 12   methodologies are very large, inadequate to ensure

 

 13   that on-line.

 

 14             The future vision that we have is that

 

 15   real-time, on-line or at-line monitoring of product

 

 16   quality can provide increased assurance of process

 

 17   in that product consistency, and that is the vision

 

 18   that we are very interested in.

 

 19             We think it reduces our risk, we think it

 

 20   improves the product quality all along the way.  We

 

 21   think the customer ultimately wants to know that

 

 22   anyway, as we do, and at the end of the day, if you

 

 23   add up the dollars, it is cost efficient, and I

 

 24   will give some examples.

 

 25             Secondly, increased process understanding

 

                                                               154

 

  1   enables risk-adjusted evaluation of process and

 

  2   product data, so that means when we do have a

 

  3   spurious event in manufacturing, which we will

 

  4   have, which everyone does have, and don't believe

 

  5   them if they tell you they are not having them, we

 

  6   can use the product quality analytical methodology

 

  7   on-line to assess the product quality impact at

 

  8   that moment and decide whether or not that batch

 

  9   should be processed or not in order to achieve a

 

 10   certain product quality attribute.

 

 11             [Slide.]

 

 12             So, let's talk about where on-line, or

 

 13   in-line, or at-line technology stands as of today.

 

 14             On this slide on your left, the three

 

 15   major stages of manufacturing include fermentation,

 

 16   purification, and formulation fill finish.

 

 17             In the second column, the purpose of each

 

 18   step certainly is to control product quality in the

 

 19   fermentation and to assure biosafety, that is, the

 

 20   adventitious agents that may impact the

 

 21   fermentation step, purification, again control

 

 22   product quality, impurity removal, ensure

 

 23   biosafety, virus clearance, bioburden clearance, et

 

 24   cetera, and finally, in formulation fill finish,

 

 25   ensure product quality, uniformity, and again

 

                                                               155

 

  1   safety.

 

  2             Present day, in the middle column, the

 

  3   third column over, in the fermentation step, we

 

  4   measure cell growth and cell viability, and we

 

  5   measure a number of metabolic parameters that, in

 

  6   part, control whether we use as control of the

 

  7   growth and the viability of the cells with the

 

  8   intent then, and the prevalidated, prospective

 

  9   validated purpose of producing the product of a

 

 10   certain quantity and a certain quality, but we do

 

 11   not measure the product quality directly in the

 

 12   cell culture step.

 

 13             In purification, again we measure process

 

 14   parameters including those listed as surrogate

 

 15   markers of control of product quality.  In order to

 

 16   measure product quality, we have to take samples

 

 17   off-line, purify the product, and measure its

 

 18   quality attributes.

 

 19             Finally, in formulation fill finish, we

 

 20   get a chance to really look at the product itself

 

 21   and the environment, the quality in terms of

 

 22   adventitious contamination and volume as an

 

 23   example, fill volume.

 

 24             So, it is not really until we get to the

 

 25   end of the process that we really get a look at the

 

                                                               156

 

  1   product quality attributes that we are trying to

 

  2   get at.

 

  3             Our view for the future then is that in

 

  4   the cell growth culture step, in the fermentation

 

  5   step, we want to be able to measure on-line,

 

  6   in-line, non-invasively, because we are trying to

 

  7   protect the fermentation from contamination, the

 

  8   content, tbe product concentration in the fermenter

 

  9   and the quality.  In fact, it could be a very

 

 10   sensitive attribute of quality.  It could be

 

 11   tertiary or quaternary structure.  It could be

 

 12   potency, it could be glycosylation.

 

 13             We want to understand the impurity profile

 

 14   and any other adventitious agents that have entered

 

 15   the step.  I want to remind you that the background

 

 16   in this step is very dirty, relatively speaking,

 

 17   that is, there are many nutrients added to the

 

 18   fermentation to promote cell growth. These are

 

 19   obviously components that we need to purify away,

 

 20   so there is a complex chemical background against

 

 21   which we are asking to measure the product quality

 

 22   and content.

 

 23             Similarly, downstream in purification, we

 

 24   want to watch quality all the time, the

 

 25   concentration as well, as we are clearing the

 

                                                               157

 

  1   impurities from the product and clearing any

 

  2   adventitious agents.

 

  3             Lastly, formulation fill finish, I think

 

  4   what we really want to do is as we are formulating

 

  5   our pre-formulation, really make sure we do have

 

  6   the right quality of the product at that point,

 

  7   because at that point in the process, the value of

 

  8   the product is very high, there has been a lot

 

  9   invested in it, and we want to make sure that we

 

 10   are going to go forward, do a fill with active,

 

 11   appropriately folded, biologically active product,

 

 12   if that is the attribute of the product at that

 

 13   stage.

 

 14             So, that gives you a vision of the future

 

 15   of what we are trying to achieve, and we feel there

 

 16   are real values to achieving that.

 

 17             [Slide.]

 

 18             Simply stated, we want to ensure that

 

 19   product quality remains consistent throughout the

 

 20   process from the beginning to the end, not just

 

 21   measuring it after the fact.

 

 22             We want to assess deviations and their

 

 23   impact in real-time, which do occur.  Some of them

 

 24   are trivial, some of them are major, nevertheless,

 

 25   the cost invested in a cell culture step is huge. 

 

                                                               158

 

  1   It's about nearly 50 percent of the total

 

  2   manufacturing cost is incurred within the cell

 

  3   culture production step.

 

  4             So, we want to avoid the cost of

 

  5   processing unreleasable batches at that stage.  So,

 

  6   this is really cost avoidance, rapid cost

 

  7   avoidance, not just ensuring product quality, we

 

  8   want to kill bad batches fast and early.

 

  9             If we want to continue processing, we have

 

 10   got the data set to justify the batch processing

 

 11   and ultimately, we will be ensured of batch

 

 12   release.

 

 13             Three.  Continual process monitoring

 

 14   obviates need for process validation.  I think that

 

 15   may be a little bit broad claim, but I think the

 

 16   implication of processing legal technology with

 

 17   regard to its impact on potential, the reductions

 

 18   in process validation is huge.

 

 19             It is huge to us because I can tell you

 

 20   today, in order to validate a process, and I am not

 

 21   just talking about the three qualification lots at

 

 22   scale prior to BLA, I am talking about all the

 

 23   process robustness studies and the assay validation

 

 24   that is done to support those process robustness

 

 25   studies.

 

                                                               159

 

  1             The industry spends 50 to 100 man-years in

 

  2   studying the impact of process perturbations on

 

  3   product quality and process yield.  That is a huge

 

  4   investment at the cost of a quarter of a million

 

  5   dollars per person year.  You do the math, it's a

 

  6   gigantic investment.

 

  7             In addition, we validate the assays that

 

  8   support the measurement of the product quality

 

  9   under those conditions.  So, there is a huge

 

 10   investment in doing process validation.  If we can

 

 11   supplant that by doing real-time process quality

 

 12   assessment and reduce process validation effort,

 

 13   that is a very big implication for the industry.

 

 14             We can reduce testing requirements at the

 

 15   end of the process potentially if we are measuring

 

 16   product quality and content all along the way.  I

 

 17   know that makes perfect sense to me as long as

 

 18   those assays are validated.

 

 19             Ultimately, we can increase process

 

 20   knowledge through identification of critical steps

 

 21   and parameters that impact quality, and this helps

 

 22   obviously improve the risk assessment and validity

 

 23   on any particular batch that is in question.

 

 24             [Slide.]

 

 25             The investment risk is substantial.  Let's

 

                                                               160

 

  1   just take on-line bioburden as an example in the

 

  2   cell culture step.  The assumptions here are

 

  3   listed.  If we are making 20 batches a year, a $20

 

  4   million annual budget, fully loaded,  that is $1

 

  5   million per batch, fully loaded, a 90 percent

 

  6   overall success rate facility, that means 18

 

  7   batches a year  gets released, that means 2 do not,

 

  8   so the cost of lost batches is $2 million a year.

 

  9             If we had on-line bioburden in the

 

 10   fermenter that could detect the contamination

 

 11   in-line, or at-line, or on-line at the time that it

 

 12   occurred, we wouldn't invest in the processing of

 

 13   that batch downstream.  That could save half the

 

 14   manufacturing costs of a batch, that is, the

 

 15   downstream costs, and we would go on with the next,

 

 16   dump that batch, and restart with the next batch.

 

 17             That is good business.  It's good business

 

 18   for the customer, it's good business for Xcellerex.

 

 19             [Slide.]

 

 20             We are involved in process analytical

 

 21   technology activities at Xcellerex.  It turns out

 

 22   we licensed the technology platform that positioned

 

 23   the company to be in this frame of thinking in the

 

 24   way of doing business, and I have listed here sort

 

 25   of four or five main bullets that we are pursuing

 

                                                               161

 

  1   at this time under process development.

 

  2             We are using high throughput screening to

 

  3   statistically optimize process parameters.

 

  4             Two.  We are using process analytics to

 

  5   look at glycosylation, for instance, and microarray

 

  6   technology, process control via noninvasive sensors

 

  7   which we have developed, including pH and DO2, and

 

  8   we are using on-line environmental monitoring of

 

  9   non-viable particulates in our manufacturing steps

 

 10   in our modular systems.

 

 11             The benefit from optimization of process

 

 12   development, on the right, is certainly to optimize

 

 13   the process from the start, to examine more

 

 14   parameters in less time.

 

 15             So, we are doing very large statistically

 

 16   designed experiments now to screen many more

 

 17   parameters that could affect product quality or

 

 18   yield.  We are using automation robotics to do

 

 19   that.

 

 20             In process analytics with these real-time

 

 21   assays, we can assess product quality in complex

 

 22   backgrounds, not in-line, but at-line at this

 

 23   point, and the non-invasive nature of the sensors

 

 24   allows us to avoid contamination of the process

 

 25   stream.

 

                                                               162

 

  1             Ultimately, on-line will bring us

 

  2   real-time assessment of environmental parameters

 

  3   and control, as I mentioned.

 

  4             [Slide.]

 

  5             So, specifically, in manufacturing--sorry,

 

  6   that was more of a focus on process development

 

  7   analytics--but in the manufacturing, what we have

 

  8   really implemented in automation include electronic

 

  9   batch records, non-invasive sensors, and on-line

 

 10   quality assurance.

 

 11             So, again, this is showing us a broader

 

 12   view of PAT, so at manufacturing level, we are not

 

 13   just talking about on-line sensors and on-line

 

 14   activity, on-line analytics, we are talking about

 

 15   an overall quality attribute or quality program

 

 16   that achieves what we think is a higher level of

 

 17   product quality, and automation is one vehicle to

 

 18   do that.

 

 19             We are using controlled environment

 

 20   modules to separate the operators in the process,

 

 21   and we are using disposables.  On the right is

 

 22   listed the benefits of doing business this way.

 

 23             So, this is our sort of approach to

 

 24   process analytical technology, but again thinking

 

 25   on the broader level of product quality knowledge

 

                                                               163

 

  1   and improvement in the manufacturing.

 

  2             [Slide.]

 

  3             With regard to knowledge management, and

 

  4   data, trending, and archiving, we have put in a

 

  5   system that is getting right at that, and so we are

 

  6   right in line with the   PAT philosophy of using

 

  7   process knowledge historically, archiving it,

 

  8   trending it, statistically analyzing it with our

 

  9   eFactory platform.

 

 10             [Slide.]

 

 11             In process optimization, here is another

 

 12   example. We do many multivariate studies with many,

 

 13   many combinations of variables.  This is a graph of

 

 14   one experiment in which we have cross-plotted the

 

 15   results of duplicates in one experiment in which we

 

 16   have looked at over 300 different media

 

 17   formulations.

 

 18             Through these methodologies, which give us

 

 19   the statistical data and power, shown in the

 

 20   numbers on the right, lower right, we really get a

 

 21   good look at process robustness and parameters that

 

 22   affect product quality and  yield.

 

 23             So, the automation and robotics puts us

 

 24   ahead in terms of understanding more about our

 

 25   process before it goes into manufacturing.

 

                                                               164

 

  1             [Slide.]

 

  2             Electronic batch records.  Here is a

 

  3   picture.  This gives us the ability to provide

 

  4   on-line quality assurance, which is again advocated

 

  5   by the PAT guidance.  We use electronic batch

 

  6   records to catch compliance issues with the

 

  7   operators, signatures, quantities, process control

 

  8   parameters, so that real-time, we are catching

 

  9   product or process control parameters that are out

 

 10   of spec or out of control.

 

 11             [Slide.]

 

 12             We look at our data historically.  Here is

 

 13   a chart of 30 batches or so, and there are four

 

 14   lines on the graph showing the data from different

 

 15   parameters that we are measuring.  In fact, this is

 

 16   a composite graph of the step yields of the

 

 17   process, so there are four steps in each graph of

 

 18   data points, is showing the step yield for that

 

 19   particular step in the process.

 

 20             The point here is that we are using

 

 21   statistical process control as advocated in the PAT

 

 22   guidance to learn about the process, to learn more

 

 23   about what affects product quality, product

 

 24   performance, and product yield.

 

 25             [Slide.]

 

                                                               165

 

  1             A couple of more slides.  I would like to

 

  2   just mention the challenges in applying process

 

  3   analytical technology to biologics.  Really, I

 

  4   think are three or four  key points.

 

  5             First, the investment in bringing

 

  6   analytics on-line is not trivial.  We don't see a

 

  7   big driver to do that, and I think that, as I

 

  8   mentioned, cost drivers will, in fact, I believe

 

  9   stimulate more investment in on-line or at-line

 

 10   technology.

 

 11             I think until we have cost pressure in the

 

 12   industry, there will not be a huge driver to do

 

 13   that.

 

 14             Secondly, innovation to develop analytical

 

 15   tools to assess critical attributes really is where

 

 16   it has to start.  It has to start back in the lab

 

 17   long before you get to the manufacturing line, you

 

 18   have got to be back in the lab converting the

 

 19   technology to something that is applicable on-line,

 

 20   perfecting that technology, miniaturizing it, and

 

 21   designing it to work in the plant floor.

 

 22             Extensive data has to be accumulated then

 

 23   in order to validate the methodology to identify

 

 24   those critical attributes and appropriate limits

 

 25   for that on-line method.

 

                                                               166

 

  1             Always, there is a regulatory uncertainty.

 

  2   I think we are always concerned about more data

 

  3   revealing more variation, and why would we want

 

  4   that if, in fact, the variation is out of spec.

 

  5   So, that is always a concern, and I think it is a

 

  6   matter of a lot of the date.

 

  7             Stringency of limits related to the

 

  8   criticality of impact gets to how widely you are

 

  9   going to validate the variance tolerance in your

 

 10   process with your on-line methodologies.

 

 11             [Slide.]

 

 12             Again, the regulatory risk is data, how

 

 13   much data is too much data, what is the collection

 

 14   interval, continuous versus intermittent data

 

 15   collection.  How to use that data, speeding release

 

 16   or speeding off-line release post-batch, or

 

 17   real-time release, and how to manage noise.

 

 18             At the end of the day, we do not want to

 

 19   lose product itself, we do not want to lose

 

 20   productivity or lower the plant output.  It just

 

 21   leads to higher manufacturing costs and lost

 

 22   product quality for the client.

 

 23             [Slide.]

 

 24             Here is an example of a continuous

 

 25   real-time data set with spurious spikes.  This

 

                                                               167

 

  1   process actually tested the in-control, but, in

 

  2   fact, we had spikes during the continuous

 

  3   monitoring, are those spikes meaningful.

 

  4             The organization needs to have a mechanism

 

  5   by which to analyze spurious spikes due to

 

  6   electronic noise or other things in order to ensure

 

  7   that it is an issue or not an issue.

 

  8             [Slide.]

 

  9             In summary, we think that the impact of

 

 10   PAT is as follows.  First, we clearly want to

 

 11   measure the product quality in the process stream,

 

 12   and we support that.

 

 13             Secondly, we want to increase the

 

 14   understanding of the process and the product

 

 15   quality relationship.  There is a relationship.  It

 

 16   is not just product by process, or process by

 

 17   product.  The two go together and understanding

 

 18   more about that is money in the bank.

 

 19             Third, continual process monitoring

 

 20   obviates the need for process validation.  We think

 

 21   that is possible within limits.

 

 22             Fourth, we believe that PAT enables

 

 23   science-based decisionmaking real-time in

 

 24   manufacturing where it has a huge value.  It can

 

 25   reduce batch release time ultimately at the end of

 

                                                               168

 

  1   the batch, and can ultimately increase plant

 

  2   capacity. It can overall lower manufacturing risk

 

  3   and, in fact, lower our cost of goods delivered.

 

  4             So, in summary, we think after all those

 

  5   things, PAT technology really can be a very cost

 

  6   effective investment for a manufacturing

 

  7   organization.

 

  8             Thank you.

 

  9             DR. KIBBE:  Thank you.  Are there any

 

 10   quick questions?  Then, we will move on to our next

 

 11   speaker.

 

 12             DR. COONEY:  Parrish, do you see any

 

 13   particular needs in the guidance that have been put

 

 14   forward so far on PAT to extend it to biologics?

 

 15             MR. GALLIHER:  Yes, we had a discussion

 

 16   actually a week or so ago with Ajaz and team.  I

 

 17   think the impact of reduction in process validation

 

 18   is understated in the guidance as written.  We

 

 19   would like to explore that further and perhaps

 

 20   expand the interpretation and the understanding of

 

 21   the impact biologics.

 

 22             The cost of process robustness study and

 

 23   the cost of validation is huge, and it may or may

 

 24   not be the right way to go ultimately if we are

 

 25   really thinking about PAT.  So, I think that is

 

                                                               169

 

  1   particularly an area for biologics that I would

 

  2   think about.

 

  3             DR. SINGPURWALLA:  I have two comments.

 

  4   One is your control charts.  It looks like your

 

  5   step one is out of control, right?

 

  6             MR. GALLIHER:  That data does not show any

 

  7   release parameters.

 

  8             DR. SINGPURWALLA:  The top one, to me it

 

  9   seems like it is out of control, but that is a

 

 10   minor point.  The major point is this, that we have

 

 11   had two talks, one by yourself, one by the previous

 

 12   speaker, and what we have seen is extolling the

 

 13   virtues of PAT into your particular industry.

 

 14             It is my sense that the FDA has taken the

 

 15   initiative and the lead in terms of infusing PAT

 

 16   into the pharmaceutical industry.  It has made you

 

 17   more efficient, presumably you saved some money.

 

 18   How much of that money has trickled down to the

 

 19   consumer as a consequence, or is there any estimate

 

 20   of that?  Because it is government investment in

 

 21   the end.

 

 22             MR. GALLIHER:  I am not sure I can answer

 

 23   that directly.  I would say that in biotech

 

 24   manufacturing, cost pressure is not really present,

 

 25   so consumer cost reduction interest in the

 

                                                               170

 

  1   pharmaceutical, at the end of the day, has not

 

  2   really trickled back to manufacturing organizations

 

  3   as part of biotech pharma companies saying to

 

  4   manufacturing you must lower costs.

 

  5             The game has been to get to market quickly

 

  6   or to the clinic and to produce enough product.  We

 

  7   have not seen on a broad scale yet the trickle-down

 

  8   of high cost of drugs, biopharmaceutical drugs to

 

  9   the manufacturing floor.  It has not really

 

 10   happened.

 

 11             That is why I said at the beginning of the

 

 12   talk, I think that is in the next decade.  As

 

 13   managed care begins to trickle back down through

 

 14   the pharmaceutical value chain to the manufacturing

 

 15   floor, we will begin to see it.

 

 16             DR. SINGPURWALLA:  So, what has been the

 

 17   gain then?

 

 18             MR. GALLIHER:  The gain for manufacturing,

 

 19   the gain for the organization?

 

 20             DR. SINGPURWALLA:  Yes.

 

 21             MR. GALLIHER:  The gain for the

 

 22   pharmaceutical organization is to reduce its

 

 23   operating costs and therefore, presumably,

 

 24   hopefully, to increase profits.

 

 25             DR. SINGPURWALLA:  Ah, but I want to see

 

                                                               171

 

  1   some of your profits come to me.

 

  2             [Laughter.]

 

  3             MR. GALLIHER:  Well, maybe we should have

 

  4   a talk outside.

 

  5             DR. KIBBE:  Anybody else?

 

  6             DR. SELASSIE:  I have a broad question on

 

  7   your statistical process control.  Are they

 

  8   sequential and are those the overall yields for the

 

  9   whole process?

 

 10             MR. GALLIHER:  This is an example of a

 

 11   process development data set, where in process

 

 12   development, again, this is where we are building

 

 13   information about the process, this is not actual

 

 14   manufacturing runs.

 

 15             We are looking at the performance of

 

 16   different steps in the process and the yield.  The

 

 17   lines that go through the data points are averages

 

 18   of the data.

 

 19             DR. SELASSIE:  I am kind of curious

 

 20   because it looks like as you go from one step to

 

 21   the fourth step, I mean the yields gradually go

 

 22   down.  Is that the overall yield or just the yield

 

 23   for each step?

 

 24             MR. GALLIHER:  Each line is the step

 

 25   yield.  Again, this is listed here as an example.

 

                                                               172

 

  1             DR. SWADENER:  Since this is a sense of an

 

  2   evaluation of the process, I am presuming that some

 

  3   of this is used to determine whether some steps are

 

  4   necessary or not in the monitoring process?

 

  5             MR. GALLIHER:  Well, what we do is we look

 

  6   at this, this is the kind of data that we look at

 

  7   to determine whether or not there are trends

 

  8   starting to impact the manufacturing controls, so

 

  9   instead of just looking at a few data points at a

 

 10   time, we look over a number of data points to

 

 11   determine if there is a trend developing in the

 

 12   data.

 

 13             We have shown this graph as a process

 

 14   development data set illustrative of the process of

 

 15   looking at many data points over a long period to

 

 16   determine if there is a trend in the data, in the

 

 17   performance of the process that you wouldn't see if

 

 18   you were just looking at a few batches at a time.

 

 19             DR. SWADENER:  Do you sometimes find that

 

 20   some of your data points that you thought were good

 

 21   data points, were not good data points, therefore,

 

 22   you don't need to use them?

 

 23             MR. GALLIHER:  No.  I mean if we are

 

 24   producing a pharmaceutical drug for intended human

 

 25   use, each batch is tested and has to meet with

 

                                                               173

 

  1   these criteria before it is ever released.

 

  2             DR. SWADENER:  But suppose one data point

 

  3   consistently comes out with the same results all

 

  4   the time, and doesn't tell you much?

 

  5             MR. GALLIHER:  Well, each assay is

 

  6   validated to ensure that it is measuring the

 

  7   intended attribute of the product, so we are quite

 

  8   sure that that is not happening.

 

  9             In those particular assays, there are

 

 10   controls that are included in those analytical

 

 11   assays to ensure that the analytical method is, in

 

 12   fact, valid every time it is run.

 

 13             DR. SWADENER:  What I am saying is suppose

 

 14   a given data point consistently comes up with the

 

 15   same result, and it is not really adding any new

 

 16   data to the whole process, can you drop that one

 

 17   and move it somewhere else?

 

 18             MR. GALLIHER:  Not without going through

 

 19   the program of change control, which is a regulated

 

 20   method of evolving process analytical technology or

 

 21   release assays or process methodologies or

 

 22   controls.

 

 23             DR. KIBBE:  I think we need to move on.

 

 24   We are gaining back all the time we saved this

 

 25   morning.

 

                                                               174

 

  1             Thank you very much.

 

  2             We have a representative from the

 

  3   pharmaceutical segment manager of Siemens Energy &

 

  4   Automation, Troy Logan.

 

  5             MR. LOGAN:  Good afternoon.  I would like

 

  6   to start by thanking the committee for providing

 

  7   the opportunity to speak here today about some of

 

  8   the experiences that Siemens has had with process

 

  9   analytical technologies.

 

 10             [Slide.]

 

 11             The PAT opportunities that are listed in

 

 12   the PAT draft guidance published by the FDA are

 

 13   that it can help to reduce production time, to have

 

 14   faster production lead time, also right first time

 

 15   quality, which means that the whole quality system

 

 16   is an integral part of the process, and a kind of

 

 17   quality system built in by design.

 

 18             Also, managing variability, trying to

 

 19   reduce the variability of the process to have a

 

 20   more consistent process.

 

 21             Facilitating continuous processing meaning

 

 22   that we can move faster from one unit operation to

 

 23   the next with fewer waiting times, which most of

 

 24   the time are due to laboratory tests.

 

 25             We can increase automation to improve

 

                                                               175

 

  1   operator safety and reduce human errors, which is

 

  2   more of a risk consideration.

 

  3             Then, the ultimate goal is real-time

 

  4   product release.  In fact, to achieve real-time

 

  5   product release, we need to achieve the first steps

 

  6   listed above.

 

  7             [Slide.]

 

  8             Real-time product release means that we

 

  9   can release the product to the market without a

 

 10   final test, so without doing laboratory tests, but

 

 11   just by reviewing process characteristics.

 

 12             [Slide.]

 

 13             If we consider the whole biopharmaceutical

 

 14   process, there are a few steps which are very

 

 15   important and which have a big impact on the

 

 16   quality of the product.

 

 17             For instance, the bioreactor stage is one

 

 18   of the most important steps because it has a large

 

 19   impact on the final quality of the product,

 

 20   compared to separation and purification where the

 

 21   quality cannot be changed very much. We can only

 

 22   isolate the desired product out of the

 

 23   fermentation.

 

 24             So, in fact, the first step where PAT

 

 25   should be applied is in steps where the impact on

 

                                                               176

 

  1   quality is the highest, and this is the bioreactor

 

  2   stage.  Later in the manufacturing process, we see

 

  3   that the biggest impact on final product quality is

 

  4   in the formulation step, so that is why formulation

 

  5   happens to be the first one addressed for PAT for

 

  6   drug manufacturing, also known as secondary

 

  7   manufacturing.

 

  8             All other areas can benefit similarly from

 

  9   PAT.  The idea is to start with the areas where it

 

 10   will have the greatest impact and the returns will

 

 11   be the greatest.

 

 12             [Slide.]

 

 13             To achieve real-time product release, we

 

 14   need to bring together many disciplines, and we

 

 15   must carefully consider the capabilities of each as

 

 16   we do.  For instance, we have to combine

 

 17   manufacturing execution systems together with

 

 18   advanced control systems, with process modeling,

 

 19   also with process development, with multivariate

 

 20   data analysis or chemometrics, with process

 

 21   understanding and with process analytics, all, of

 

 22   course, inside of a regulatory framework.

 

 23             [Slide.]

 

 24             If we look at the whole concept, there is

 

 25   the process layer on the bottom and the IT

 

                                                               177

 

  1   infrastructure on the top.  There are two aspects

 

  2   of this whole PAT concept, the control aspect on

 

  3   one side and the process monitoring aspect on the

 

  4   other.

 

  5             Looking inside the boxes, we see that the

 

  6   control solution is built out of control modules

 

  7   and equipment modules, brought together to form

 

  8   pharmaceutical modules, a batch management system,

 

  9   and, of course, electronic batch records, which are

 

 10   fed into the MES or IT infrastructure.

 

 11             On the other side are the process

 

 12   analytics which  can be applied in two ways.

 

 13   First, for process specification verification and

 

 14   real-time product release, and, second, for

 

 15   collecting information from the process to apply an

 

 16   iterative learning control system that will help to

 

 17   increase our knowledge of the process on the fly as

 

 18   the process runs and, based on that, improve the

 

 19   control strategy.

 

 20             Further on top, as you gain more knowledge

 

 21   about your process, you can begin to optimize that

 

 22   process.

 

 23             [Slide.]

 

 24             We look now to a real world example, that

 

 25   is,  control of a bioreactor, which is typically

 

                                                               178

 

  1   based on monitoring pH, dissolved oxygen and

 

  2   temperature, and apply a closed loop control

 

  3   strategy based on the information from these

 

  4   sensors.

 

  5             If we now also introduce a PAT solution,

 

  6   it can help by providing more information about the

 

  7   process, not just secondary parameters, but also

 

  8   chemical composition and biological performance or

 

  9   biological status of the process. This information

 

 10   can then be used as an input to the control system.

 

 11             Separate from this, there is typically a

 

 12   laboratory that is checking the quality of the

 

 13   product and making decisions about holding or

 

 14   releasing the product to the market.

 

 15             A future strategy can be that decisions

 

 16   are no longer made in the laboratory, but instead,

 

 17   the process control system on the manufacturing

 

 18   floor decides, based on the information obtained

 

 19   from PAT, if product will be held or released to

 

 20   the market.

 

 21             [Slide.]

 

 22             This is an example of where we have

 

 23   applied PAT for fast identification of

 

 24   contaminations or a certain disturbance in a

 

 25   process.  This is from a yeast-based fermentation

 

                                                               179

 

  1   where the major threat to the process is

 

  2   contamination introduced by microorganisms coming

 

  3   in through air that is sparged into the bioreactor

 

  4   or via the substrate before it is transferred into

 

  5   the bioreactor.

 

  6               The conventional laboratory test

 

  7   normally take 8 hours before it is known if this

 

  8   contamination has taken place.  With this new way

 

  9   of applying PAT, we are able to quickly, within a

 

 10   few seconds, identify when there is a

 

 11   contamination.

 

 12             [Slide.]

 

 13             What you see here is a representation of

 

 14   this kind of classification.  On purpose, we have

 

 15   contaminated the yeast fermentation with 7 of the

 

 16   most common microorganisms that, in the case of

 

 17   this company, caused one-third of their rejected

 

 18   batches, so that means significant economic impact

 

 19   in their business.

 

 20             We intentionally contaminated the

 

 21   fermentation and found that we can classify and

 

 22   identify the outcome into contaminated or not

 

 23   contaminated product, and this chart is the result

 

 24   of that experiment.

 

 25             [Slide.]

 

                                                               180

 

  1             Here is another example.  An in-situ probe

 

  2   was placed inside a bioreactor and is monitoring

 

  3   the process by collecting the spectra from the

 

  4   beginning to the end, and a principal component

 

  5   analysis is being applied.  The principal component

 

  6   analysis is used to monitor process change

 

  7   throughout the batch.

 

  8             [Slide.]

 

  9             What you see here is a two principal

 

 10   component plot that represents the major changes of

 

 11   this process.  From a process control point of

 

 12   view, we are mainly interested in what is changing

 

 13   in the process, so we would like for everything

 

 14   that is staying constant to be taken out of what is

 

 15   being monitored.  That is exactly what a principal

 

 16   component analysis does.

 

 17             The result of this principal component

 

 18   analysis is a plot that is called a process

 

 19   fingerprint.  It represents a typical batch track.

 

 20   The next step is to define the ideal track, which

 

 21   is the so-called "golden" track.  By following this

 

 22   track, the required endproduct quality can be

 

 23   achieved.

 

 24             The next step is to determine the maximum

 

 25   acceptable tolerance to achieve the required

 

                                                               181

 

  1   endproduct quality.  That is this tunnel, which can

 

  2   be calculated based on good batches.  This is a

 

  3   kind of standard deviation that we will allow

 

  4   around the process track.

 

  5             This is a great tool for helping to define

 

  6   if the process is running consistently.  Also, if

 

  7   sudden process disturbances occur, it is a fast

 

  8   detection tool that helps to avoid lasting impact

 

  9   of those disturbances.

 

 10             Shown here in the middle of this chart is

 

 11   where we had a disturbance due to an oxygen

 

 12   depletion when an oxygen valve was blocked.

 

 13             [Slide.]

 

 14             Now we have the PAT road map

 

 15   implementation stages for the implementation of

 

 16   PAT.  It consists of three major steps.  First, is

 

 17   the measuring part including monitoring and process

 

 18   understanding.  The second one is the controls, and

 

 19   the third is optimization.

 

 20             Along with these three steps we have some

 

 21   parallel tracks.  One is knowledge and change

 

 22   management, another is the validation aspect, and

 

 23   the third, the people and organizational issues.

 

 24   Because the introduction of this PAT solution will

 

 25   cause a lot of changes in the organization, people

 

                                                               182

 

  1   have to make decisions differently.

 

  2             For example, the decision on holding or

 

  3   releasing product will be made on the manufacturing

 

  4   floor and no longer in the laboratory.   This means

 

  5   that the work processes of the organization must be

 

  6   realigned.

 

  7             We start first with risk assessment on

 

  8   product quality and on the process, so to determine

 

  9   the required product quality and assess the process

 

 10   to determine which process parameters are the

 

 11   relevant ones to track.

 

 12             The third part is the analyzer assessment,

 

 13   finding out which analyzer is most appropriate for

 

 14   the type of process and what information is needed

 

 15   from that analyzer. Once all of that information is

 

 16   collected, a multivariate data analysis is

 

 17   conducted.  This focuses on finding the

 

 18   relationships between product quality and process

 

 19   parameters.

 

 20             Based on that, you can then begin the

 

 21   design of experiments.  The PAT solution will then

 

 22   begin to help to determine which are the good

 

 23   batches and isolate the "golden" batch.

 

 24             The next step is then control.  Here, we

 

 25   can modify control parameters if the process goes

 

                                                               183

 

  1   off track, and we can get an understanding of the

 

  2   ideal process control strategy.  When the process

 

  3   is running off track, other techniques can be used

 

  4   to get the process under control again, and we can

 

  5   improve process knowledge with the application of

 

  6   an iterative learning control strategy.

 

  7             During all these different steps, we are

 

  8   collecting a lot of information - process behavior,

 

  9   process capabilities, process quality, et cetera.

 

 10             This data can then be used to further

 

 11   optimize the process meaning we can further

 

 12   optimize the "golden" process track, perhaps the

 

 13   processing time can be shortened, improving

 

 14   efficiency of equipment utilization, or the process

 

 15   can be optimized to use fewer resources and still

 

 16   achieve the required final endproduct quality.

 

 17              In conclusion, the use of these PAT

 

 18   technologies will become part of an ongoing

 

 19   strategy of continuous process improvement.

 

 20             Thank you for your attention.

 

 21             DR. KIBBE:  Quick questions, anyone?

 

 22             DR. SINGPURWALLA:  Your second slide said

 

 23   something about production release of

 

 24   pharmaceuticals without final tests.

 

 25             MR. LOGAN:  Yes.

 

                                                               184

 

  1             DR. SINGPURWALLA:  Are you serious about

 

  2   that?

 

  3             MR. LOGAN:  I will have to begin by

 

  4   answering that I am really here as a spokesperson

 

  5   for our technical people, but as I understand it,

 

  6   that is the ultimate end goal that they are

 

  7   attempting to achieve, and they are seriously

 

  8   pursuing it with the end users that we are trying

 

  9   to work with.

 

 10             DR. SINGPURWALLA:  Maybe that needs a

 

 11   point of clarification.  You don't want to test,

 

 12   you cannot test every product because if you tested

 

 13   it, you couldn't sell it.  When I buy a pill, it is

 

 14   presumably not tested, but  then you still want to

 

 15   sample even though you use PAT techniques at the

 

 16   end, you do want to sample.

 

 17             Here is an analogy.  Suppose you are

 

 18   building an airplane engine, it has got many parts.

 

 19   You test each part. There is no guarantee that when

 

 20   you put it all together, the engine will function.

 

 21   So, you still need to do testing at the end to make

 

 22   sure that nothing has been overlooked.  No?  Why is

 

 23   that?

 

 24             DR. KIBBE:  I will allow your colleague to

 

 25   respond.  We will let you off the hook.

 

                                                               185

 

  1             Anybody else have anything?

 

  2             [No response.]

 

  3             DR. KIBBE:  Thank you very much.

 

  4             Our last speaker during the open public

 

  5   hearing is someone from Laboratory Instrumentation

 

  6   Scientist, Foss-NIRSystems, Robert Mattes.

 

  7             MR. MATTES:  Thank you.  I am Robert

 

  8   Mattes.

 

  9             I would like to talk to you today about

 

 10   near infrared spectroscopy as possibly one of these

 

 11   analytic tools that would help in the toolbox for

 

 12   PAT, and by demonstrating some of our experiences

 

 13   in PAT so far as we have implemented some

 

 14   techniques in the tableting arena.

 

 15             [Slide.]

 

 16             The near infrared, just so everybody

 

 17   knows, is the region between the visible and the

 

 18   mid-IR, and it looks at overtones of the

 

 19   fundamental absorptions in the mid-IR.

 

 20             [Slide.]

 

 21             One of the things that we have done for

 

 22   years using near infrared has been the inspection

 

 23   of incoming raw materials, and we can measure them

 

 24   for identification and qualification of those

 

 25   materials, so that you are making sure that you

 

                                                               186

 

  1   have the right materials going into a process

 

  2   fermentation cell before starting a reaction.

 

  3             One of the things we also have had a lot

 

  4   of experience in and has been implemented in

 

  5   manufacturing environments is measurement of

 

  6   moisture content and lyophilized product.  I am

 

  7   going to show some data from each of those.

 

  8             [Slide.]

 

  9             I have a similar chart to the last speaker

 

 10   here that shows, first of all, the typical types of

 

 11   monitoring that we do real time In a process

 

 12   reactor for temperature, pH, oxygen level, you

 

 13   know, and you are controlling the temperature and

 

 14   sparging and pH level.

 

 15             With the near-infrared probe also

 

 16   introduced directly into the process reactor, we

 

 17   are able to measure analytes, amino acids, glucose

 

 18   levels, feedstock levels in that process reactor

 

 19   real time, which helps the manufacturing people a

 

 20   great deal.

 

 21             We haven't actually installed this in, in

 

 22   research laboratories and plants so far.

 

 23             So, if you are looking at the raw

 

 24   materials that we have now identified and qualified

 

 25   being brought into the bioreactor, then, we are

 

                                                               187

 

  1   monitoring the analytes and can real-time adjust pH

 

  2   or nutrient levels, and so forth, according to the

 

  3   data that we get.

 

  4             As the product comes out, we can measure

 

  5   the moisture content as the product is being dried

 

  6   and possibly lyophilized.  This can also lead to

 

  7   control feedback for process improvement through

 

  8   statistical process control charts, and so forth,

 

  9   as previous people have mentioned.

 

 10             [Slide.]

 

 11             Here are some of the organisms that we

 

 12   have worked with, Escherichia coli, products like

 

 13   you see on the list there, and the biomass also.

 

 14   With one spectrum that you take in the near

 

 15   infrared, you can analyze multiple components

 

 16   instantaneously with the same spectrum.

 

 17             In fact, we are working on one experiment

 

 18   right now where we are looking at 28 different

 

 19   analytes including all the amino acids, glucose,

 

 20   glutamate, lactate, and so forth.  Really, your

 

 21   requirements are not limited in that sense.

 

 22             [Slide.]

 

 23             Here is an example of a process, the raw

 

 24   near-infrared spectra of a process as it

 

 25   progresses.  As biomass increases the y axis

 

                                                               188

 

  1   spectra or the absorbance of the spectra increases,

 

  2   as you can see here.

 

  3             [Slide.]

 

  4             You see these two major peaks are the

 

  5   water bands in the near infrared.  It is not

 

  6   terribly informative in that form.  We usually take

 

  7   the second derivative of the spectra, which

 

  8   enhances the resolution and enhances the peak

 

  9   separations.

 

 10             In the top set of spectra here, we see one

 

 11   analyte progressing with time, and it is increasing

 

 12   in a downward direction because of that second

 

 13   derivative that we have taken.  In the lower set of

 

 14   spectra, we see where different analytes are

 

 15   appearing within a process.

 

 16             [Slide.]

 

 17             The colors on the charts are backwards in

 

 18   the overhead here, but I have corrected them in

 

 19   your handout, I am sorry about that, but the

 

 20   biomass should be in red as you will see it

 

 21   increasing with time in the process, and the

 

 22   glycerol content was decreasing there.

 

 23             So, you can see using those spectra, you

 

 24   can predict and measure the levels of different

 

 25   analytes and trend them with time rather than

 

                                                               189

 

  1   waiting for a week or more sometimes, waiting for

 

  2   the wet chemistry to come back on a process that

 

  3   you are running presently.

 

  4             [Slide.]

 

  5             Here are some of the types of things and

 

  6   the types of error in precision that we have been

 

  7   able to develop.  At-line, we are talking about

 

  8   using a peristaltic pump that pumps out of the

 

  9   reactor and back in again.  In-line, we are talking

 

 10   about actually having it pulled right in the

 

 11   reactor, which is the type of work I have been

 

 12   working on most recently with some of our

 

 13   customers.

 

 14             [Slide.]

 

 15             One of the things that has been reported

 

 16   recently  in biotechnology and bioengineering by a

 

 17   group that worked at Strathclyde University in the

 

 18   UK was CHO cell fermentation, which is a very big

 

 19   topic right now.

 

 20             They used a small, 2-liter bioreactor

 

 21   similar to the one you saw in a previous lecture

 

 22   there, and they were monitoring glucose, glutamine,

 

 23   lactate, and ammonia.

 

 24             [Slide.]

 

 25             This is more like the work that I was

 

                                                               190

 

  1   doing most presently with about a 100-liter

 

  2   bioreactor with a direct fermenter interface, using

 

  3   standard Ingold port.  We are putting our probe

 

  4   right into the sterile environment.  The probe can

 

  5   be sterilized right in the environment.

 

  6             As I say, we simultaneously can get

 

  7   results in less than one minute of up to 28

 

  8   analytes.  Previously, we have to have developed

 

  9   the model for each one of those.  The

 

 10   time-consuming part is upfront on the analysis

 

 11   rather than the real-time usually used in wet

 

 12   chemistry.

 

 13             This then can be turned into monitoring

 

 14   and closed-loop control, adding feed or whatever,

 

 15   changing glucose levels real-time automatically,

 

 16   but certainly in the nearest future, will help the

 

 17   people to know when the levels have changed to a

 

 18   serious level within a reactor real-time.

 

 19             [Slide.]

 

 20             Here is the results of that particular

 

 21   experiment with the CHO cells.  You can see the

 

 22   precisions and ranges that were used in that

 

 23   experiment with ammonia, glucose, lactate, and

 

 24   glutamine.

 

 25             [Slide.]

 

                                                               191

 

  1             Here is an example of monitoring

 

  2   lyophilized product.  You mentioned earlier that

 

  3   not every sample can be measured or would be

 

  4   destroyed.  In this case, we can, we actually

 

  5   non-invasively, non-destructively measure right

 

  6   through the bottom of the lyophilized bottle, and

 

  7   we can predict the moisture content.  So, there is

 

  8   a possibility of 100 percent measurement in this

 

  9   case.

 

 10             You see this band, the largest band there

 

 11   is the water band.  Again, it's the second

 

 12   derivative, so it is increasing in downward

 

 13   direction.  The driest bottle would be the red line

 

 14   that is up at the top.

 

 15             [Slide.]

 

 16             Some of the benefits for the PAT

 

 17   initiative in the biotech area.  It gives a

 

 18   real-time analysis of sterile environments.  You

 

 19   don't have to constantly be taking samples out that

 

 20   could lead to problems with sterility and asepsis

 

 21   and also the possibility of closed-loop feedback

 

 22   process control.

 

 23             It is not invasive and can add to the

 

 24   process optimization, as people spoke of earlier,

 

 25   waste reduction, and better understanding of your

 

                                                               192

 

  1   process, so that they can create safer and

 

  2   improved, more consistent product.

 

  3             Thank you.  Are there any questions?

 

  4             DR. KIBBE:  Questions?

 

  5             DR. SINGPURWALLA:  This is very

 

  6   interesting.  So, this is a non-invasive method of

 

  7   looking at some particular unit, but then do you

 

  8   have a template for what would be a normal unit,

 

  9   and how do you compare these templates?

 

 10             Suppose you have a template which says

 

 11   this is what the spectrum of a proper product

 

 12   should be, and then you get a defective, how do you

 

 13   say this is defective?

 

 14             MR. MATTES:  What you are talking about is

 

 15   a qualitative analysis before predicting the sample

 

 16   quantitatively?  Yes, we can build libraries, and I

 

 17   have done quite a bit of work like this recently.

 

 18             You want to build a library of what

 

 19   qualified good samples of spectra should look like,

 

 20   and if it doesn't conform to those criteria,

 

 21   statistical criteria that you have developed in

 

 22   your library, it gives you some sort of indication,

 

 23   or it will not give you a prediction as such, so

 

 24   you won't be predicting on the wrong type of

 

 25   spectrum.

 

                                                               193

 

  1             DR. SINGPURWALLA:  What you need is a

 

  2   template which measures the spectrum of a good

 

  3   product versus the spectrum of a defective product

 

  4   and the criteria for seeing how diverse those two

 

  5   are, because, you know, a little diversity, you

 

  6   cannot say it's bad or good, but you need proper

 

  7   criteria to say that this is very diverse, and I

 

  8   don't know if you have that, but it is interesting.

 

  9             MR. MATTES:  Yes.  We use statistical

 

 10   criteria in our library model developments, and we

 

 11   can use bad samples as reject sets, so we can test

 

 12   both positive and negative sets, and it is

 

 13   basically, to use the simplest example, if you had

 

 14   normal distributions of 3-sigma outlier or you

 

 15   choose some number of standard deviations from the

 

 16   mean-centered spectrum of this acceptable

 

 17   population.

 

 18             DR. KIBBE:  Anybody else?

 

 19             DR. COONEY:  An extension of the previous

 

 20   question.  You are using the sensor to measure

 

 21   multiple components in variable and complex

 

 22   systems.  To what extent do you have to go back and

 

 23   redevelop the algorithm for each system for the

 

 24   components versus being able to use standard

 

 25   wavelengths or a template, as was asked, that you

 

                                                               194

 

  1   can apply across different processes?

 

  2             MR. MATTES:  Well, each unique process

 

  3   really needs the model development done for that

 

  4   process, so this, as I say, is the upfront

 

  5   time-consuming portion of this model development,

 

  6   but you simultaneously are looking at all the

 

  7   variance caused by all the different constituents

 

  8   or analytes in the matrix of your fermentation.

 

  9             So you need many samples, reference

 

 10   samples, to help you be able to do this, because

 

 11   you are going to have so many degrees of freedom,

 

 12   you need more samples.

 

 13             DR. KIBBE:  Thank you.  The table at the

 

 14   back end is just references?

 

 15             MR. MATTES:  Yes, it is just a

 

 16   bibliography that has some references including the

 

 17   work that Strathclyde University did on the CHO

 

 18   cell mammalian culture.

 

 19             DR. KIBBE:  Great.  Thank you very much.

 

 20             Now we are back to the PAT Applications

 

 21   for Products in the Office of Biotechnology

 

 22   Products, and we are going to start off with Keith

 

 23   Webber.  Keith is here ready to lead the charge.

 

 24               PAT Applications for Products in the

 

 25                 Office of Biotechnology Products

 

                                                               195

 

  1                       Overview and Issues

 

  2              DR. WEBBER:  Good afternoon.  I am Keith

 

  3   Webber and I would like to thank the committee for

 

  4   taking the time today to participate and listen to

 

  5   the issues surrounding our desire to implement PAT

 

  6   technologies for the products in the Office of

 

  7   Biotechnology Products.

 

  8             As Ajaz mentioned this morning, the PAT

 

  9   guidance specifically excluded the biotech

 

 10   products, that are regulated in our office, from

 

 11   its scope.

 

 12             To some extent, this was to expedite the

 

 13   publication of the document and also the training

 

 14   and qualification program for inspectors and

 

 15   reviewers, but as he also said, it is a technology

 

 16   that is certainly amenable to any manufacturing

 

 17   process, so there is not inherently any reason why

 

 18   we couldn't implement it with these products if we

 

 19   have the technologies and the information and

 

 20   understanding available.

 

 21             [Slide.]

 

 22             Now, this afternoon, just to give you a

 

 23   brief overview of the agenda here, I am going to

 

 24   give an overview basically of the biotech products

 

 25   and the manufacturing processes for the products

 

                                                               196

 

  1   that are regulated in our office, and then Dr.

 

  2   Joneckis from CBER will give a brief overview

 

  3   related to some of the products that are regulated

 

  4   in CBER.

 

  5             After that, two members of the committee,

 

  6   Dr. Cooney and Dr. Koch, will give presentations to

 

  7   describe some of the issues, as well as some of the

 

  8   opportunities available in the area of fermentation

 

  9   and biological manufacturing.

 

 10             That will be followed by Dr. Layloff, who

 

 11   will give a brief overview of the view in this area

 

 12   with regard to the PAT Subcommittee which he

 

 13   chaired when it was active.

 

 14             Afterwards, we will put up some questions

 

 15   to stimulate discussion.  I certainly hope that we

 

 16   will get a good amount of discussion from the

 

 17   committee with regard to this exciting area of

 

 18   manufacturing.

 

 19             [Slide.]

 

 20             The biological products as a class include

 

 21   all the products listed here, which were originally

 

 22   regulated in CBER.  There was a reorganization back

 

 23   in 2003 that moved the recombinant DNA-derived

 

 24   proteins, or many of them I should say, to the

 

 25   newly formed Office of Biotechnology Products

 

                                                               197

 

  1   within the Office of Pharmaceutical Sciences in

 

  2   CDER.

 

  3             Essentially, those are the products that I

 

  4   am going to be focusing on today.  Dr. Joneckis may

 

  5   have comments on the other products, as well, or

 

  6   the recombinant DNA products that are still

 

  7   remaining in CBER.

 

  8             [Slide.]

 

  9             This is in terms of sort of a review.

 

 10             [Slide.]

 

 11             There are essentially two aspects of

 

 12   process analytical technologies.  One requirement

 

 13   is that you have to have the ability to monitor the

 

 14   critical product characteristics that are needed

 

 15   for the product's function, or, if it is an

 

 16   intermediate in manufacturing, you need to be able

 

 17   to know what characteristics are important for

 

 18   being able to move it forward in manufacturing to

 

 19   the next step.

 

 20             Now, alternatively, there may be

 

 21   surrogates as opposed to direct product quality

 

 22   attributes that one can use to make decisions.

 

 23   This monitoring, as has been mentioned a number of

 

 24   times here, will optimally be done on-line, but at

 

 25   this point, I think to a large extent, many of the

 

                                                               198

 

  1   monitoring is done off-line, so this is something

 

  2   we look for in the future.

 

  3             Secondly, one has to be able to monitor

 

  4   and modulate the critical process parameters to be

 

  5   able to guide the product quality attributes and

 

  6   quality characteristics during the manufacturing

 

  7   process.

 

  8             It is probably worth mentioning two other

 

  9   requirements that may be self-evident, but are

 

 10   certainly not trivial, that is, that you need to

 

 11   know the critical characteristics of the product in

 

 12   the first place that are important for its function

 

 13   or that need to be obtained to get to the next step

 

 14   in manufacturing.

 

 15             You also need to know how these

 

 16   characteristics can be modified and manipulated by

 

 17   the manufacturing process parameters themselves.

 

 18   That is one area that is really dependent upon

 

 19   industry to determine during their period of

 

 20   product development and gaining a thorough

 

 21   understanding of their product and their process.

 

 22             [Slide.]

 

 23             This is really part of a come-down version

 

 24   of process analytical technologies, but I think has

 

 25   most of the important aspects with regard to the

 

                                                               199

 

  1   manufacturing element itself.  One has a process,

 

  2   unit operation, one is monitoring the process

 

  3   characteristics or process parameters, as well as,

 

  4   if possible, the product characteristics during the

 

  5   process.

 

  6             You gather this data, evaluate it, and

 

  7   then make decisions, so that one can adjust the

 

  8   process to ensure that the product that is coming

 

  9   out of that process is going to have the

 

 10   appropriate characteristics that are desirable.

 

 11             [Slide.]

 

 12             This is just a brief overview, which we

 

 13   have seen already in one of the earlier

 

 14   presentations, of the          various

 

 15   biotechnology processes that are utilized.  This

 

 16   isn't all-inclusive, but are the major ones.

 

 17             You have fermentations, harvesting from

 

 18   the fermenter.  You have product capture from that

 

 19   harvest.  Concentration is usually a step that goes

 

 20   on after, and may be a part of product capture.

 

 21             There are filtrations that are done often,

 

 22   almost always chromatography of some sort, many

 

 23   times multiple steps.  There is formulation

 

 24   process, and if the products are lyophilized

 

 25   products, you then have lyophilization process at

 

                                                               200

 

  1   the end.

 

  2             I didn't cover filling operations, but

 

  3   those are certainly amenable to PAT, as well.

 

  4             [Slide.]

 

  5             Now, what are the characteristics of the

 

  6   biotech APIs that are generally considered to be

 

  7   critical quality attributes?  Certainly, the

 

  8   primary amino acid sequence is critical to the

 

  9   proper functioning of the product, however, this is

 

 10   a characteristic that is relatively invariant,  I

 

 11   would say, once you get into the manufacturing

 

 12   area, and it is established at the master cell bank

 

 13   stage or the working cell bank stage, so it is

 

 14   usually not looked at on a lot-to-lot basis.

 

 15             The secondary structure pertains to the

 

 16   local interactions between the amino acid residues

 

 17   to produce a structure, such as the alpha helix,

 

 18   the pink you see in the front, and the beta pleated

 

 19   sheets that you see in the back, in yellow.

 

 20             The secondary structure is really very

 

 21   important to the protein because these are the

 

 22   structures that serve as the building blocks to

 

 23   produce enzymatically active sites or the binding

 

 24   sites for protein.

 

 25             [Slide.]

 

                                                               201

 

  1             They come together, as I mentioned, to

 

  2   form tertiary structures.  This is illustrated here

 

  3   in this figure by a model of an antibody FAV

 

  4   fragment.  You can see that this is purely beta

 

  5   pleated sheet, and these tertiary structures form

 

  6   to form the binding sites of the antibody itself.

 

  7             The next level of complexity that is

 

  8   characteristic of some proteins is the assembly of

 

  9   independent protein molecules into multimeric

 

 10   quaternary structures.  Such structures assemble

 

 11   post-translationally and they are generally held

 

 12   together by either ionic or hydrophobic

 

 13   interactions between the independent subunits.

 

 14             [Slide.]

 

 15             The last, but not least certainly, of the

 

 16   API characteristics that I am going to talk about

 

 17   today are the post-translational modifications.

 

 18   Glycosylation is probably one of the most common

 

 19   post-translational modifications that is of concern

 

 20   with proteins, particularly those that are made in

 

 21   the eukaryotic cells.

 

 22             It is illustrated in this figure by the

 

 23   sugar chains that are in the center of the Fc

 

 24   fragment of an antibody molecule.

 

 25              Glycosylation patterns and structures are

 

                                                               202

 

  1   highly variable in proteins from one product to the

 

  2   next, and they can be significantly altered, as

 

  3   mentioned earlier, by the fermentation conditions

 

  4   that occur during cell growth and fermentation.

 

  5             The other modifications that are seen in

 

  6   proteins include the proteolytic cleavages that can

 

  7   either be caused by endoproteinases that chew away

 

  8   at one end of the molecule or exoproteinases--I am

 

  9   sorry, endoproteinases that eat the middle--may be

 

 10   producing the final product as a necessary activity

 

 11   to get the product you want, or the exoproteinases

 

 12   which eat away at the end of the protein and could

 

 13   produce degradation products during the

 

 14   manufacturing process.

 

 15             There also is often or sometimes you see

 

 16   acylations and sulfations, and many other

 

 17   post-translational modifications that I really

 

 18   won't describe here.

 

 19             [Slide.]

 

 20             Now, leaving API on its own and looking at

 

 21   the product characteristics themselves, which

 

 22   really then you get into the whole impurity profile

 

 23   of the product and  excipients that may be present.

 

 24              Impurities fall into two categories, the

 

 25   process-related impurities, which are media

 

                                                               203

 

  1   components coming from the fermentation process,

 

  2   host cell proteins that would come from the

 

  3   expression system, and then leachates, which come

 

  4   from columns or containers that are used to store

 

  5   the product during processing.

 

  6             Then, also, you have product-related

 

  7   impurities, which are perhaps truncations of the

 

  8   molecules or misfolded molecules or aggregates of

 

  9   the product, which can occur during storage or even

 

 10   during manufacturing.

 

 11             [Slide.]

 

 12             Now, I would like to discuss briefly some

 

 13   of the analytical methods that are used currently

 

 14   to look at these factors for biotech products or

 

 15   these characteristics of biotech products.

 

 16             As was mentioned earlier, the primary

 

 17   structure is really something that is not looked on

 

 18   at a lot-to-lot basis unless in particular cases,

 

 19   you might have, as I mentioned, a cleavage of a

 

 20   protein that is part of the manufacturing process.

 

 21   In those cases, then, one generally does look at

 

 22   the primary structure, not necessarily with

 

 23   sequencing, but just to demonstrate that cleavage

 

 24   has occurred appropriately.

 

 25             One area that I also would note here, for

 

                                                               204

 

  1   products that are patient-specific products, for

 

  2   example, antibodies that are used for treating

 

  3   B-cell lymphomas where each individual patient gets

 

  4   a unique product.  There is an area where the

 

  5   primary structure would certainly be critical to

 

  6   look at as an identity test, if nothing else, prior

 

  7   to giving a product to the patients.

 

  8             [Slide.]

 

  9             The secondary structure is somewhat more

 

 10   difficult to evaluate, and that is because there is

 

 11   a limited number of direct techniques.  The ones

 

 12   that are primarily used are circular dichroism and

 

 13   NMR at this point.

 

 14             Also, another complicating factor for

 

 15   proteins is that most proteins have multiple

 

 16   secondary structures in them.  For antibodies, it

 

 17   is almost all beta-pleated sheet, but other

 

 18   proteins, you have a mixture, so you need to have a

 

 19   method that will be able either to distill out the

 

 20   critical values for that protein or can look at the

 

 21   individual secondary structures separately.

 

 22             One other complicating factor for this

 

 23   with regard to an in-process control, which we

 

 24   hopefully will be able to overcome at some point,

 

 25   is they need relatively pure material to look at

 

                                                               205

 

  1   secondary structures in a protein.

 

  2             [Slide.]

 

  3             Now, I grouped the tertiary and quaternary

 

  4   structures together because they are both high

 

  5   order structures and are amenable to a similar set

 

  6   of analytical tools.

 

  7             The functional assays, such as in-vitro

 

  8   potency assays, can directly measure the

 

  9   therapeutic--or I shouldn't say the

 

 10   therapeutic--but the activity of the product

 

 11   itself, so it is semi-looked upon as a surrogate,

 

 12   but actually, it is a measure usually of the direct

 

 13   activity, but it requires, of course, the

 

 14   product-specific reagents to do that.

 

 15             This is also true of the immunoassays.

 

 16   You can get a direct picture of the structure of

 

 17   the protein if you have antibodies that will bind

 

 18   to 3-dimensional epitopes that are relevant to the

 

 19   tertiary or quaternary structure, but again you

 

 20   need to have product-specific reagents to do that.

 

 21             Peptide mapping is a valuable method for

 

 22   looking at the disulfide bonds to make sure that

 

 23   they are mapped, that they are forming

 

 24   appropriately.

 

 25             Size-exclusion chromatography is a

 

                                                               206

 

  1   relatively insensitive method for looking at

 

  2   tertiary structure, but in some cases, you can use

 

  3   it to separate monomeric from multimeric forms of

 

  4   the protein, so that can be a very useful

 

  5   technique.

 

  6             Hydrophobic-interaction chromatography is

 

  7   actually a very good method because it looks at the

 

  8   surface charges and surface characteristics of the

 

  9   protein and can be used to very sensitively detect

 

 10   either misfolded proteins or proteins that are not

 

 11   associated with their other monomers appropriately.

 

 12             [Slide.]

 

 13             For post-translational modifications, this

 

 14   is probably the most variable characteristic of the

 

 15   protein, as I mentioned before, and analyses of

 

 16   these usually requires a highly purified protein

 

 17   and some rather sophisticated methodologies, for

 

 18   example, enzymatic cleavage and analysis of the

 

 19   amino-linked oligosaccharide protein, however,

 

 20   recently, the mass spec and NMR have allowed direct

 

 21   analysis of post-translational modifications in

 

 22   intact proteins, which is an up and coming

 

 23   technique.

 

 24             Peptide mapping can also pinpoint the

 

 25   location of the modification within the protein

 

                                                               207

 

  1   sequence, which is very useful for characterization

 

  2   of the product.

 

  3             Immunoassays and the functional assays can

 

  4   be used for more impure proteins because they are a

 

  5   little bit more specific for your product, however,

 

  6   the functional assays are often not very sensitive

 

  7   to protein modification itself unless there is a

 

  8   specific modification that is really critical to

 

  9   the activity.

 

 10             [Slide.]

 

 11             So, to summarize, inherent challenges that

 

 12   we see to implementing PAT for biotech products at

 

 13   this point are that the biotech products are

 

 14   generally large and complex pleiotropic molecules.

 

 15             They are composed usually of a mixture of

 

 16   post-translational modifications, they have

 

 17   multiple active sites.  Some of those are

 

 18   homologous like two binding sites antibody, or they

 

 19   can be heterologous where you have different active

 

 20   sites doing different functions on the same

 

 21   protein.

 

 22             The activities are dependent upon the

 

 23   complex, folded conformations of a protein, and

 

 24   proteins are also susceptible to multiple

 

 25   degradative events, so you need to look at a lot of

 

                                                               208

 

  1   different aspects of a protein during

 

  2   manufacturing.  As I mentioned before, these

 

  3   include the proteolysis, aggregation, misfolding,

 

  4   oxidation, deamidation, just to name some of those

 

  5   that we know of.

 

  6             [Slide.]

 

  7             Of course, when you are considering the

 

  8   factors involved in protein structure or actually

 

  9   any product, you need to consider the purity,

 

 10   potency, and the strength, of course, but also the

 

 11   impact that those changes or modifications or

 

 12   variabilities to the protein would have on the

 

 13   pharmacokinetics, the pharmacodynamics, and the

 

 14   immunogenicity of the product.

 

 15             That is delving more into the area of the

 

 16   product development stage of pharmaceutical

 

 17   development as opposed to manufacturing itself, but

 

 18   surely, that is one of the early bits of

 

 19   information that one needs to have, we need to

 

 20   gather.

 

 21             [Slide.]

 

 22             Now, I would like to talk briefly about a

 

 23   few of the manufacturing processes that have been

 

 24   touched on before and what the current state of

 

 25   monitor and control are.

 

                                                               209

 

  1             For fermentation processes, generally, one

 

  2   can monitor and control the agitation rate, the pH,

 

  3   the ionic strength of the media, the temperature,

 

  4   dissolved gases, media components, and by being

 

  5   able to monitor and control those, you can then

 

  6   control the growth rate and the expression rate

 

  7   usually of your product.

 

  8             This is an area where process analytical

 

  9   technology, we will probably see it developed

 

 10   early, because one has that control over some of

 

 11   the aspects of the process.

 

 12             As we have heard before, there are methods

 

 13   now available for detecting or monitoring the

 

 14   biomass and bioburden through using rapid

 

 15   biological methods, rapid microbiological methods

 

 16   for sterility testing.  Generally, one monitors the

 

 17   product by light absorbance, for example, protein

 

 18   concentration to A280.

 

 19             [Slide.]

 

 20             Moving on to chromatographic processes,

 

 21   this is again the same format.  You can monitor and

 

 22   control your pH of the effluent or the liquid

 

 23   phase, ionic strength, flow rate, temperature, and

 

 24   volume, and of value here, which isn't exactly laid

 

 25   out, though, is that because you can control the

 

                                                               210

 

  1   volume and monitor the light absorbance, one can

 

  2   then control the composition to some extent of the

 

  3   fractions that you collect out of that, from that

 

  4   column.

 

  5             That is currently being done although it

 

  6   is really looking just at the protein

 

  7   concentration, one usually doesn't know except by

 

  8   doing previous experiments, to know what is in each

 

  9   of the fractions that you collect.

 

 10             [Slide.]

 

 11             Filtration processes.  This includes both

 

 12   dead-end filtrations for removal of bacteria and

 

 13   viruses, as well as the ultra-filtration for

 

 14   selectively removing lower and higher molecular

 

 15   species from a product.

 

 16             In most cases, one can monitor and control

 

 17   the temperature and flow rate, the back pressure,

 

 18   and the volume of the filtrate, although you

 

 19   usually can't do all those independently because

 

 20   they are inter-related.

 

 21             Again, we have seen before the protein

 

 22   concentration is monitored by light absorbance and

 

 23   the bioburden is, at this point, generally

 

 24   monitored off-line, but soon could be monitored

 

 25   on-line.

 

                                                               211

 

  1             Dead-end filtration is usually a

 

  2   flow-through process and that generally allows

 

  3   little control over the product characteristics

 

  4   themselves other than the removal of the material

 

  5   which is filtered out.

 

  6             Ultra-filtration, on the other hand, can

 

  7   be a much more dynamic process, and that may allow

 

  8   more control over the composition of the product.

 

  9   For example, ultra-filtration is often used for

 

 10   formulation of biotech products.

 

 11             [Slide.]

 

 12             It was discussed a little bit earlier, the

 

 13   lyophilization process, and this one may be

 

 14   currently the most close to being a process

 

 15   analytical technology.  In the lyophilizer, you can

 

 16   monitor and control the shelf temperature and the

 

 17   product temperature, the chamber pressure, the

 

 18   condenser temperature, the pressure, and time in

 

 19   the lyophilizer.

 

 20             The ability to monitor and control these

 

 21   parameters allows you to control the freezing rate

 

 22   and the drying rate, and the moisture content, all

 

 23   of which directly affect the physical quality of

 

 24   the final product, which really is what we are

 

 25   shooting for in process analytical technologies.

 

                                                               212

 

  1             Although you have to have a product with

 

  2   an acceptable composition going into the

 

  3   lyophilizer, the physical characteristics of the

 

  4   product that comes out will play an important role

 

  5   in the stability and the activity of the product

 

  6   that goes to the patient.

 

  7             [Slide.]

 

  8             Finally, you will see these questions

 

  9   again at the end of our session, but I just want to

 

 10   introduce them now, because these are points that

 

 11   we would like to initiate discussion with.

 

 12             What technologies are available now to

 

 13   evaluate the characteristics of protein products in

 

 14   real time during manufacturing, or to speed things

 

 15   along with an off-line test which is faster, is

 

 16   valuable to know, as well.

 

 17             What tools would allow us to understand

 

 18   the manufacturing process better?

 

 19             What processes in biological drug

 

 20   manufacturing would benefit the most from

 

 21   implementation of PAT?  Essentially, where are we

 

 22   going to get the most bang for our buck, as has

 

 23   been said before.

 

 24             For processes or products that do not

 

 25   currently allow direct product quality monitoring,

 

                                                               213

 

  1   what other strategies would you, as a committee,

 

  2   recommend for product quality control in addition

 

  3   to control of the in-process parameters?

 

  4             Finally, what additional elements should

 

  5   be incorporated in a training and certification

 

  6   program for reviewers and inspectors of

 

  7   biotechnology PAT applications?

 

  8             Thank you.

 

  9             DR. KIBBE:  Does anybody have any

 

 10   questions?  It might be a good idea for us to go

 

 11   ahead and get at least the next speaker through the

 

 12   process, and I think it might be useful for the

 

 13   committee to be able to take a break then, so I

 

 14   don't know how that does to your continuity, but it

 

 15   would be helpful for us.

 

 16             DR. JONECKIS:  Thank you and good

 

 17   afternoon.  I am Chris Joneckis.  I am the Senior

 

 18   Adviser for CMC Issues in CBER, Office of the

 

 19   Director.

 

 20             I am just going to briefly describe CBER's

 

 21   perspective on process analytical technologies for

 

 22   the biotechnology and biological products that CBER

 

 23   currently regulates.

 

 24             [Slide.]

 

 25             CBER regulates a wide variety of products,

 

                                                               214

 

  1   as shown in these slides, the majority of the major

 

  2   product classes shown here.  They include a wide

 

  3   variety of biological and biotechnology products,

 

  4   diagnostic and processing devices, cells, and even

 

  5   chemical entities that are clearly derived from a

 

  6   variety of sources and manufactured using a wide

 

  7   variety of techniques.

 

  8             My comments today will predominantly focus

 

  9   on the experience that we have gained with the more

 

 10   traditional biologics and some of the newer

 

 11   recombinant products that are produced from living

 

 12   organisms and are typically extracted and further

 

 13   modified, purified, and, for example, fill for

 

 14   distribution following some of the examples that

 

 15   Keith provided in the manufacturing process.

 

 16             For many of these products, most actually,

 

 17   product contamination with adventitious agents from

 

 18   a variety of sources is of primary concern, and

 

 19   most of these products are again aseptically

 

 20   processed.

 

 21             It is important to point out also that

 

 22   there are recombinant products not just in the

 

 23   blood derivative class for the recombinant

 

 24   analogues that CBER regulates, but also in a

 

 25   variety of other classes including allergenic

 

                                                               215

 

  1   extracts, prophylactic and therapeutic vaccines.

 

  2   They are also used in the manufacture of various

 

  3   cellular therapies and in some other product

 

  4   classes not shown here.

 

  5             CBER's approach to technology in general

 

  6   is also applicable to other product classes, and

 

  7   many of the comments on PAT that I will make today

 

  8   will be applicable to those.

 

  9             [Slide.]

 

 10             Historically, CBER's approach to

 

 11   controlling the process can clearly be summed up by

 

 12   the mantra, if you will, that, "The process is the

 

 13   product."

 

 14             There has been a long historical emphasis

 

 15   in understanding the product and a long emphasis on

 

 16   understanding and controlling that manufacturing

 

 17   process. This clearly requires, not just an

 

 18   understanding of the process and the product, but

 

 19   the interaction of those two, how the process

 

 20   results in the product.

 

 21             The nature of many of the traditional

 

 22   biologics influenced this approach.  Many of these

 

 23   were complex heterogeneous products susceptible to

 

 24   a variety of variability produced almost

 

 25   exclusively from living sources or living sources

 

                                                               216

 

  1   themselves.

 

  2             The complex mixtures, coupled with

 

  3   insufficient analytical technologies, made it very

 

  4   difficult to detect all the active components or

 

  5   materials, in fact, that can influence the activity

 

  6   of the active components.

 

  7             This necessitated a very strict control of

 

  8   the manufacturing process to reproducibly result in

 

  9   the desired product with the appropriate safety and

 

 10   efficacy profile.

 

 11             Recent advances in analytical technology

 

 12   and enhanced manufacturing processes often result

 

 13   in better defined products, aiding in a greater

 

 14   assurance of producing products with the desired

 

 15   characteristics.

 

 16             Manufacturing is beneficial to implement

 

 17   these newer technologies and improved approaches to

 

 18   better control processes and demonstrate that

 

 19   products can be consistently manufactured.  That

 

 20   was clearly shown in many of the recently derived

 

 21   biotechnology and biological products.

 

 22             [Slide.]

 

 23             An overall approach that we have followed

 

 24   at CBER has been that we have always encouraged the

 

 25   application of technologies and concepts to the

 

                                                               217

 

  1   manufacturing and testing of products.

 

  2             Again, we have lived with developing

 

  3   technologies throughout its history, and have

 

  4   applied those to manufacturing and testing of

 

  5   various products.  We are actively involved in the

 

  6   development and application of these new

 

  7   technologies.

 

  8             Again, historically, we have developed and

 

  9   applied technologies appropriate to specific

 

 10   manufacturing and testing issues.  We continue to

 

 11   be actively engaged in developing and applying

 

 12   these technologies.

 

 13             For example, the conversion of older

 

 14   technologically-based assays, such as animal-based

 

 15   assays and cell-based assays to newer analytical

 

 16   methods, actively involved again in development and

 

 17   application of proteonomics and genomic

 

 18   technologies to issues, such as product

 

 19   characterization and adventitious agent detection.

 

 20             This large laboratory component assists us

 

 21   in maintaining our knowledge base for discussions

 

 22   in applying these new technologies.

 

 23             We clearly partner with manufacturers in

 

 24   developing and implementing new technologies and

 

 25   concepts. As I have indicated, we have had to live

 

                                                               218

 

  1   with developing technology throughout history.

 

  2             It is through these interactions with the

 

  3   manufacturers in both development and in

 

  4   post-approval phases that allowed the advancement

 

  5   and development and introduction of new

 

  6   technologies or appropriate manufacturing

 

  7   processes.

 

  8             Issues are addressed, validation issues,

 

  9   for example, and other types of issues about

 

 10   understanding this new technology are addressed

 

 11   throughout the development process, the

 

 12   post-approval process, on review and inspection, as

 

 13   well as in review of applications.

 

 14             [Slide.]

 

 15             The approach to process control that CBER

 

 16   has emphasized is best described as a comprehensive

 

 17   life-cycle approach to validate this process and

 

 18   spans the life cycle of that product.

 

 19             This approach relies on developing an

 

 20   understanding of the process and product.  Use of

 

 21   knowledge gained can be applied throughout the life

 

 22   cycle and typically is.

 

 23             In addition to CBER's perspective, this

 

 24   comprehensive approach was largely influenced

 

 25   through interactions with manufacturers of

 

                                                               219

 

  1   biologics and biotechnology products, incorporated

 

  2   concepts and approaches often used in manufacturing

 

  3   industries.

 

  4             It emphasizes identification and control

 

  5   of critical unit operations and process variables

 

  6   to product intermediates, resulting in a product

 

  7   with acceptable quality attributes.

 

  8             Some of the elements are shown here.  They

 

  9   are familiar I am sure to many of you.  They also,

 

 10   I should point out, overlap with many of the

 

 11   fundamental underlying principles necessary to

 

 12   implement many of the PAT applications.

 

 13             [Slide.]

 

 14             As a result, over time, there have been

 

 15   many PAT-like applications of technology to

 

 16   manufacturing and testing.  For example, as Keith

 

 17   had indicated, there are many examples of continual

 

 18   on-line monitoring of critical process attributes

 

 19   often with real-time feedback mechanisms that may

 

 20   be computer assisted.

 

 21             Within the defined parameters from the

 

 22   validation studies and such there is also some

 

 23   flexible control within those parameters, so one is

 

 24   not necessarily fixed to certain endpoints if the

 

 25   appropriate validation characteristics support a

 

                                                               220

 

  1   range within which one can operate.

 

  2             We have been involved with application of

 

  3   on-line analysis of various intermediates and

 

  4   product attributes, as well as facility systems.

 

  5   Some examples are indicated here.

 

  6             We have approved several years ago an

 

  7   on-line measure of a critical physical-chemical

 

  8   quality intermediate for a naturally-derived

 

  9   product.  We have entertained discussions, again

 

 10   several years ago, on measuring through a

 

 11   non-destructive method the moisture content of

 

 12   final filled containers.  We have approved

 

 13   appropriate physical property for changing

 

 14   lyophilization conditions in lyophilizers.

 

 15             CBER regulates and reviews major facility

 

 16   changes. We approve numerous supplements that

 

 17   described on-line applications of water systems

 

 18   when conductivity measurements were substituted for

 

 19   the wet chemistry measurements in water systems.

 

 20             Most importantly, we have recently

 

 21   approved microbial methods for two applications.

 

 22   Rapid microbial methods are very concerned

 

 23   especially to or for our cellular products, and for

 

 24   those products where they cannot be held or stored

 

 25   prior to the release of the sterility testing

 

                                                               221

 

  1   results, so implementing methods that have allowed

 

  2   for a rapid turnaround to determine whether the

 

  3   products are sterile or not has provided great

 

  4   increase in the assurance of the quality of that

 

  5   product.

 

  6             I should point out that that last method

 

  7   is not an on-line method, but is an off-line

 

  8   method.

 

  9             [Slide.]

 

 10             There are clearly potential applications

 

 11   for new manufacturing and testing technologies that

 

 12   have been discussed.  Many of those advantages for

 

 13   PAT have been described and are probably known much

 

 14   better to you all than to me.

 

 15             I think some of the best applications

 

 16   would be if one could use those in terms of

 

 17   defining product or intermediate quality

 

 18   characteristics.  Unfortunately, that provides the

 

 19   most challenge and at present, I think there are

 

 20   some great limitations to doing that in an on-line

 

 21   fashion.

 

 22             Immediate applications I think may be more

 

 23   likely in terms of drug product manufacturing,

 

 24   measuring of more single types of process or other

 

 25   very select quality attributes.

 

                                                               222

 

  1             Some of the challenges I think that we

 

  2   face at CBER is that we still have, in contrast to

 

  3   some of the more purified and defined recombinant

 

  4   products, a large amount of complex and

 

  5   heterogeneous products.  All of the issues that

 

  6   Keith discussed about the identity, purity, and

 

  7   composition of these products is in many cases

 

  8   magnified when one has a complex and heterogeneous

 

  9   product.

 

 10             Again, I think that leads to the ability

 

 11   that it may be difficult to know from a multifactor

 

 12   analysis the heterogeneous mixture, what actually

 

 13   that relationship is.

 

 14             Again, manufacturing unit operations in

 

 15   biological and biotechnology products often perform

 

 16   multiple functions. Again, the ability to measure

 

 17   all important product quality characteristics in a

 

 18   continuous mode from any of those functions, I

 

 19   think is going to be very challenging.

 

 20             For CBER, we have the development of new

 

 21   products, not just products within a class, but

 

 22   again completely new products, gene therapy,

 

 23   therapeutic vaccines, as well as cellular products.

 

 24             [Slide.]

 

 25             Just in summary, I think it is still

 

                                                               223

 

  1   important at CBER that we understand and emphasize,

 

  2   understanding both the product and the process,

 

  3   clearly integral to the development and manufacture

 

  4   of biotechnology and biological products.

 

  5             The comprehensive, life-cycle approach to

 

  6   process validation remains integral to the

 

  7   consistent manufacture of these products.

 

  8   Validation is still a regulatory requirement and

 

  9   when conducted in a comprehensive life-cycle

 

 10   manner, has provided great assurance that the

 

 11   process will consistently produce that desired

 

 12   product.

 

 13             That has been most readily seen at CBER

 

 14   when products that were approved prior to

 

 15   validation being a regulatory requirement,

 

 16   validated their process.  They had potential

 

 17   savings both from economic and public health

 

 18   perspectives.

 

 19             We see PAT more as an extension of the

 

 20   existing process understanding the manufacturing

 

 21   control paradigm.  I think clearly, PAT has

 

 22   potential applications for biotechnology and

 

 23   biological manufacturing processes especially if it

 

 24   can monitor again intermediate quality attributes

 

 25   and provide greater assurance of that product

 

                                                               224

 

  1   quality.

 

  2             We will continue to partner with

 

  3   manufacturers of existing and new products to

 

  4   facilitate implementing any type of new technology

 

  5   and concepts, including those that can enhance the

 

  6   knowledge and control of the manufacturing process.

 

  7             Thank you.

 

  8             DR. KIBBE:  Does anybody have any

 

  9   questions for Chris?  Go ahead.

 

 10             DR. COONEY:  One of the particular

 

 11   challenges for the class of products you are

 

 12   dealing with are viruses, viral contamination.

 

 13             How do you see some of the issues of

 

 14   detection and validation of viral removal being

 

 15   advanced by PAT?

 

 16             DR. JONECKIS:  That is an interesting

 

 17   question. Currently, I guess, for the committee's

 

 18   benefit, most people do challenge or clearance

 

 19   studies, usually small scale, representative of the

 

 20   larger scale manufacturing process.

 

 21             In terms of detection, again, as I

 

 22   mentioned earlier, there are efforts underway to do

 

 23   genomic and proteomic screenings for potential

 

 24   contaminants within products at various appropriate

 

 25   stages in addition to the current various levels of

 

                                                               225

 

  1   safety that are provided.

 

  2             I suppose theoretically if one could with

 

  3   the increased sensitivity of certain methods, one

 

  4   may be able to do more on-line monitoring, if you

 

  5   would, again at early or appropriate stages to

 

  6   actually see if there is any type of potential

 

  7   viral materials present.

 

  8             One could potentially in theory, depending

 

  9   upon how much is present, again, sensitivity of

 

 10   your methods, actually measure on-line for the

 

 11   various steps, present of type C retroviral

 

 12   particles, CHO-derived products, and things of that

 

 13   nature.

 

 14             Similarly, you know, it has been done for

 

 15   measuring DNA and other types of materials when it

 

 16   is there in a large amount in early purification

 

 17   steps, given the sensitivity of the assay, one can

 

 18   measure those on-line in addition to whatever model

 

 19   studies are done to provide additional assurance

 

 20   that your model truly reflects what is occurring.

 

 21             DR. KIBBE:  Anyone else?

 

 22             Seeing none, I am going to take the

 

 23   prerogative of the Chair and declare a 15-minute

 

 24   break, which means we should be back in our seats

 

 25   and ready to go at approximately 2:35.

 

                                                               226

 

  1             [Break.]

 

  2             DR. KIBBE:  I have been assured by experts

 

  3   in the field that Tom has all the answers in his

 

  4   presentation, so when we get to them, we will be

 

  5   done for the day.

 

  6             Charles Cooney is on the podium.

 

  7             DR. COONEY:  Thank you very much.

 

  8             I am pleased to have an opportunity to

 

  9   share some thoughts this afternoon on the question

 

 10   that Keith Webber put before us, and that is the

 

 11   extension of PAT to biological processes.

 

 12             In preparing for any talk, one obsesses

 

 13   over a number of things, one of which is the color

 

 14   of your tie, of course, but another is the title of

 

 15   the talk.  I obsessed over a complex title and a

 

 16   simple title, and I resolved that dilemma by having

 

 17   both.

 

 18             [Slide.]

 

 19             PET for PAT?  The message that I am trying

 

 20   to convey in my title as a place to begin is that

 

 21   when we think about PAT and all of its virtues and

 

 22   aspects that have been dealt with earlier today,

 

 23   process analytical technologies applied to

 

 24   processes and products, it is a very important

 

 25   fundamental concept, and it means a lot.

 

                                                               227

 

  1             When you think about it in terms of the

 

  2   process, it occurred to me that we really need to

 

  3   think about analyzing the process, as well as

 

  4   analyzing parts of the process and the product

 

  5   itself.

 

  6             So, the emphasis here is to think about

 

  7   process evaluation tools as a component of process

 

  8   analytical technologies, and I think that the broad

 

  9   definition that has been used for PAT very much

 

 10   embraces that idea.

 

 11             [Slide.]

 

 12             In putting together my comments for this

 

 13   afternoon, I have identified more questions than I

 

 14   have answers, and the reason for this is that as we

 

 15   think about going forward with the extension of PAT

 

 16   to biological products, there are a number of

 

 17   issues and questions, and I would like to try to

 

 18   put at least a few of these into some context.

 

 19             The first set of questions I have

 

 20   summarized here as Some Issues.  What are the

 

 21   issues, what is the context as we look forward, one

 

 22   of which is the pipeline of new products, what will

 

 23   that look like going forward in the next 10 to 20

 

 24   years.

 

 25             I think there is no doubt that it is going

 

                                                               228

 

  1   to be expansive, there is going to be increased

 

  2   complexity in the nature of the products, and it is

 

  3   going to be a very vibrant pipeline simply based

 

  4   upon what we see in discovery and what we see in

 

  5   clinical trials today.

 

  6             If we think about the increase in the

 

  7   number of BLAs and NDAs that will be coming through

 

  8   for biological products, it puts a real future

 

  9   stress on the Agency because as we look at the

 

 10   number of these products, they are increasing

 

 11   exponentially, and I don't think that the number of

 

 12   people in the FDA is increasing exponentially.

 

 13   Just a guess, but I think it's true.

 

 14             So, what that means is that the pressure,

 

 15   in order to be more efficient, and to focus on a

 

 16   risk-based strategy and understand where and when

 

 17   to look, at what, is really very, very timely to be

 

 18   in this process right now.

 

 19             Then, of course, there is the question of

 

 20   follow-on biologics that are beginning to--I will

 

 21   come back to this in a moment--but are beginning to

 

 22   come forward, and I think are going to be an

 

 23   increasing issue.

 

 24             Both of these issues raise the question

 

 25   how do biological products respond to the physical

 

                                                               229

 

  1   process changes that occur when you develop a

 

  2   process, scale the process, move it, change its

 

  3   location, and the like.

 

  4             We have some understanding of this, and,

 

  5   of course, this is fundamental to understanding how

 

  6   biological products, particularly the complex one,

 

  7   respond to the complex processes used to make them.

 

  8             Underlying all this, do we have the

 

  9   adequate analytics to address the uncertainties

 

 10   associated with manufacturing in this industry, and

 

 11   I am struck by looking at the presentations we had

 

 12   earlier today, and, of course, they all focused on

 

 13   where we have the analytics in place.

 

 14             In fact, do we have the necessary

 

 15   analytics?  No, I don't think we do.

 

 16             Are efforts underway to develop them?

 

 17   Well, we are going to hear in the next presentation

 

 18   that there are some very exciting efforts that are

 

 19   underway, and I think the future looks bright, but

 

 20   it is only going to come with a lot of diligence

 

 21   and a lot of innovation in order to measure the

 

 22   kinds of things that we really need to be looking

 

 23   at.

 

 24             Then, ultimately, how do we bring this

 

 25   together to assure robustness in design and

 

                                                               230

 

  1   operation of these processes.

 

  2             [Slide.]

 

  3             I tried to address where we are going.

 

  4   Keith Webber already identified a number, in fact,

 

  5   the previous two speakers identified the range of

 

  6   products that are out there today and the ones that

 

  7   are likely to be out there tomorrow, and we can

 

  8   expect that there are going to be a lot more

 

  9   antibodies, replacement proteins, designer

 

 10   proteins, vaccines, not just for therapeutic use,

 

 11   but for prophylactic use, cellular and gene

 

 12   therapies are being developed quite aggressively.

 

 13             One of the other observations I would like

 

 14   to make, though, when we look at the range of

 

 15   products that are there today and that are going to

 

 16   be there tomorrow, is that this question of

 

 17   follow-on biologics is on the minds of many people

 

 18   and we need to take stock of where we are today,

 

 19   because we really have follow-on biologics today,

 

 20   we have multiple processes for the same products,

 

 21   multiple manufacturers for human growth hormone,

 

 22   multiple manufacturers by very diverse technologies

 

 23   for human insulin.

 

 24             How we have managed them is perhaps not

 

 25   the same way that we wish to manage them in the

 

                                                               231

 

  1   future, but these are realities today, these are

 

  2   not things that are looming out there for the

 

  3   future.

 

  4             [Slide.]

 

  5             When we look at the processes that are

 

  6   going to be used, we have very diverse recombinant

 

  7   protein production processes.  Why are there so

 

  8   many?  Why isn't there a single technology that has

 

  9   emerged?

 

 10             The answer is very simple, not all

 

 11   processes are suitable for all products.

 

 12   Furthermore, the intellectual property landscape is

 

 13   such that it dictates complexity in the processes

 

 14   that are used simply to work your way through the

 

 15   minefield of intellectual property that is out

 

 16   there.

 

 17             Is that going to get simpler as we look

 

 18   forward? No, the processes are going to become more

 

 19   complex, driven in part by innovation, and driven

 

 20   in part by the nature of the products, tissue

 

 21   products, multicellular products, and certainly the

 

 22   potential future for transgenic plants and animals.

 

 23             So, as we look at the array of complex

 

 24   processes for these complex products, I do not see

 

 25   that landscape getting simpler.  I see it remaining

 

                                                               232

 

  1   complex and as a consequence, we need to be able to

 

  2   have the analytics in place and the ways of

 

  3   handling the data and the ways of understanding

 

  4   these processes that is better in the future than

 

  5   it is today.

 

  6             So, this leads us to a series of

 

  7   challenges, and I have tried to organize these

 

  8   challenges in a way that represents where we are

 

  9   coming from and where we are going to go.

 

 10             There is the continuing challenge of

 

 11   rapid, cost effective development and scale-up.  We

 

 12   need to shorten the timelines, the timelines for

 

 13   developing the processes, and if we develop better

 

 14   processes, that should lead to improved timelines

 

 15   for approval of those processes, and we need to be

 

 16   able to have more flexibility, so that the process

 

 17   of development and scale-up could be a lot more

 

 18   nimble and lean than it is today.

 

 19             But then once we have processes in place,

 

 20   I think the industry has done an increasingly good

 

 21   job in the drug space of continuous improvement,

 

 22   and most recently, and we have heard examples of

 

 23   that today, PAT is a major contributor to how that

 

 24   is going to go forward in the future.  That is very

 

 25   positive.

 

                                                               233

 

  1             We need to understand how to better

 

  2   achieve continuous improvement in process change in

 

  3   the biological space, and that is the challenge

 

  4   that we are focusing on in this particular session.

 

  5             What the tools that we need to do that?

 

  6   What are the methodologies?  Where is the

 

  7   uncertainty, and, of course, how do we understand

 

  8   that risk, and risk is implicit in all of this.

 

  9             Follow-on biologics present their own

 

 10   challenges, and then when we get into complex

 

 11   biologicals, cellular therapies, and tissue

 

 12   engineering, there are a wide variety of unknowns

 

 13   and we need to understand quickly what are the

 

 14   parameters, what are the biomarkers, what are the

 

 15   surrogate markers, what are the direct methods that

 

 16   we can apply in order to get a grasp of these

 

 17   processes and how they will define the products

 

 18   that we make.

 

 19             Furthermore, as we look at these

 

 20   challenges, there is a constant tension between the

 

 21   safety and the economic agenda, and where is the

 

 22   proper balance in terms of how much risk we seek to

 

 23   minimize and how much risk we seek to embrace and

 

 24   manage and take forward.

 

 25             [Slide.]

 

                                                               234

 

  1             Well, when you look at the broad issue of

 

  2   the relationship between the process and the

 

  3   product, one has to look at what goes in and what

 

  4   comes out.  We have raw materials and environmental

 

  5   conditions that are variables going in.  We are

 

  6   trying to control a number of the parameters in

 

  7   this space.

 

  8             Some of those parameters are suitable for

 

  9   control in a closed loop fashion.  Again, we heard

 

 10   a number of examples of how that is increasingly

 

 11   important today.  A number of those parameters we

 

 12   don't control in a closed loop manner, but we need

 

 13   to control them nonetheless.

 

 14             I think the challenge in looking at this

 

 15   very microscopic view of a process is the

 

 16   information flow.  We know how to do process

 

 17   control.  We are going to get better at

 

 18   implementing new analytics on these processes.

 

 19   There is a long history of applying statistical

 

 20   process control and a wide variety of other

 

 21   methodologies of process control.

 

 22             We are going to get better at doing that,

 

 23   and that is all going to be incremental.  What is

 

 24   not going to be incremental is the more systems

 

 25   view of understanding to do it better.  I think

 

                                                               235

 

  1   there we will have some big jumps, but where I

 

  2   think we are doing a terrible job is on the

 

  3   information flow.

 

  4             The information is quite an asset, a lot

 

  5   of money goes into generating that information, and

 

  6   do we adequately understand and mine it, and the

 

  7   answer is no, we don't.  In fact, it's a very

 

  8   poorly utilized asset, and in some cases, the

 

  9   reason is, well, if I don't look at it, I don't

 

 10   have to worry about the variance in it.  That is

 

 11   one way to control variance.

 

 12             Another way is to say, well, let me

 

 13   embrace that variance, let me learn from it, let me

 

 14   capture that information, and feed that back and

 

 15   learn, and that is an area where I think we are

 

 16   getting better, but, frankly, I think if I look

 

 17   back over the past decade or two, even looking at

 

 18   work that I have done, I think we have done a

 

 19   pretty bad job.

 

 20             Now, what I would like to do is to stay in

 

 21   the frame of raising questions rather than

 

 22   providing answers, but I can't go through a

 

 23   presentation like this without showing some data

 

 24   and without taking an example to illustrate where I

 

 25   think there are some opportunities and some of the

 

                                                               236

 

  1   kind of learning that represents work-in-progress.

 

  2             In biological processes, one of the main

 

  3   issues we deal with is the oxygen dilemma.

 

  4             [Slide.]

 

  5             We all know that in most biological

 

  6   processes, there is a requirement for oxygen for

 

  7   efficient growth, and in this particular case,

 

  8   recombinant protein expression.  That is a given.

 

  9             By the way, there is some interesting data

 

 10   to suggest that that is not necessarily true, but

 

 11   we won't go there now.  But that is a general

 

 12   methodological given, and let's assume that it's

 

 13   true for the moment.

 

 14             But on the negative side, there is the

 

 15   potential that if a little bit of oxygen is good,

 

 16   is a lot of oxygen better, and the answer is not

 

 17   necessarily, because there is potential for both in

 

 18   vivo and in vitro protein oxidation of methionine,

 

 19   cysteines, for instance, and as we scale-up and as

 

 20   we change the amount of oxygen, as we use enriched

 

 21   oxygen in processes, is this going to be a hazard,

 

 22   is it going to be a problem?

 

 23             We wanted to explore that, and we also

 

 24   know that oxygen can induce stress.  Actually,

 

 25   oxygen too high or too low can induce stress.  One

 

                                                               237

 

  1   of my hobbies is high altitude mountaineering, and

 

  2   I decided what would it be like to operate under 35

 

  3   percent partial pressure of oxygen.  Well, I don't

 

  4   recommend going there on a regular basis.

 

  5             [Slide.]

 

  6             But when we look at processes today, we

 

  7   are looking at scale.  Traditionally, what we have

 

  8   done is to do a lot of our optimization of a

 

  9   process at a shake flask scale, 100-milliliter,

 

 10   perhaps to a 10-liter scale, and then go to 10 or

 

 11   100 cubic meter scale.

 

 12             The benefits of doing research at the

 

 13   homogeneous milliliter or liter scale is that we

 

 14   can make the assumption that it is almost

 

 15   homogeneous, and the work we have done over the

 

 16   years is to better resolve events in time, so we

 

 17   have taken analytics, like some of the probes, and

 

 18   so on, that have been discussed earlier, and we

 

 19   have learned to evolve events in time and

 

 20   understand how the time space is critical.

 

 21             At the fermentation scale, we might do 200

 

 22   to 300 experiments in order to get what we think is

 

 23   an optimum, but we really know it is not, in order

 

 24   to scale to the 10 to 100 cubic meter scale, but

 

 25   all we do is get to a place that allows us to

 

                                                               238

 

  1   economically be in the business, and then,

 

  2   hopefully, we will be allowed to undergo continuous

 

  3   improvement following that.

 

  4             What I want to suggest is that what we

 

  5   really need to think about is how we look at this

 

  6   process development and scale-up paradigm very

 

  7   differently.

 

  8             That is, if we scale down, and, for

 

  9   instance, one approach is to use reactors that are

 

 10   100 microliters, and they indeed are homogeneous,

 

 11   or somewhere in that small space, and do large

 

 12   numbers of experiments, and not just resolve events

 

 13   in time, but do the kind of things that were

 

 14   described earlier, create large experimental

 

 15   design, so that we can now not just look at our

 

 16   experimental space, but we can look at the

 

 17   interdependencies between the independent variables

 

 18   in a much more effective way, reduce the

 

 19   uncertainty associated with how the process

 

 20   responds to the environment, as well as changes

 

 21   with time, and reduce the uncertainty of scale-up,

 

 22   and presumably reduce the variance as we do so.

 

 23             That is not to say that we shouldn't also,

 

 24   at scale, resolve events that take place in time.

 

 25   There is going to be variance in a biological

 

                                                               239

 

  1   process.  We can learn a lot from that, and that

 

  2   allows us to manage the risk associated with these

 

  3   processes, and that goes on, as well, but we are

 

  4   doing a better job with that than we are simply

 

  5   going to the large-scale experimental design.

 

  6             [Slide.]

 

  7             A model system that we happened to choose

 

  8   is alpha-1 antitrypsin.  It is a human recombinant

 

  9   protein.  It is an interesting model because you

 

 10   notice that methionine 358 and the one at 351, it

 

 11   sticks up like a sore thumb and is sensitive to

 

 12   oxygen.  So, we reasoned it would be useful as a

 

 13   molecular probe in order to determine if oxidation

 

 14   was a problem.

 

 15             This molecule also actually has 10

 

 16   methionines, several of which are partially or

 

 17   completely exposed, and 1 unpaired cysteine that is

 

 18   partially exposed, but with models such as this,

 

 19   this might be the product where its structure is

 

 20   well known, you can begin to do microscale

 

 21   experiments that you can then project to the larger

 

 22   scale and ask, well, what is the effect of oxygen

 

 23   on the molecule.

 

 24             [Slide.]

 

 25             In this particular case, we observed that

 

                                                               240

 

  1   there was an oxygen-dependent proteolytic cleavage,

 

  2   and as you look at these three lines, the green

 

  3   line is for the expression, transient expression

 

  4   under air.  The top line is transient expression

 

  5   under anaerobic conditions, which turns out to be

 

  6   not so bad.  But the bottom line is expression

 

  7   under pure oxygen, so there is this oxygen

 

  8   dependency of the proteolytic cleavage.

 

  9             [Slide.]

 

 10             How do we resolve that?  Well, one

 

 11   approach is the very hypothesis-driven problem,

 

 12   and, of course, when you have a problem, and a

 

 13   complex problem, and as you can see by the photo on

 

 14   the left, if you don't get the ropes right, you

 

 15   could be in serious trouble, so you have got to

 

 16   know where the problem is if you want to be in the

 

 17   position on the right.

 

 18             [Slide.]

 

 19             So, how do we resolve that?  Well, we have

 

 20   a hypothesis.  In this case, we tried many

 

 21   hypotheses.  I am only going to tell you about the

 

 22   one that is right.  That way you will remember that

 

 23   I got it right the first time.  Wrong, but

 

 24   nonetheless, we speculated that it was the protease

 

 25   ClpP that was responsible.

 

                                                               241

 

  1             [Slide.]

 

  2             It is a complex protease that involves

 

  3   ATP.  You do the hypothesis-driven experiments, you

 

  4   knock it out, and as you can see by the figure on

 

  5   the righthand side, you eliminate the

 

  6   oxygen-dependent proteolytic cleavage, not all the

 

  7   cleavage, but that hypothesis, which was one of

 

  8   about a dozen that we explored, in fact, worked.

 

  9             [Slide.]

 

 10             Are there other ways to think about these

 

 11   kind of problems?  Do we have analytical techniques

 

 12   that allow us to probe much more broadly the global

 

 13   cell response?

 

 14             [Slide.]

 

 15             Of course, the answer is yes, and the

 

 16   technology of using DNA microarrays to do

 

 17   transcriptional profiling is one kind of tool that

 

 18   can be used in identifying where the problem is,

 

 19   and, after all, isn't that what PAT is about.

 

 20             It is about getting at the underlying

 

 21   science to understand what the issue is, and then

 

 22   focus on the right issue, not necessarily measuring

 

 23   everything that you possibly can measure.

 

 24             E. coli is very convenient.  It only has

 

 25   about 4,000 genes, but fortunately, those genes are

 

                                                               242

 

  1   set up in pathways, and rather than think about

 

  2   4,000, I don't like big numbers, I would rather

 

  3   think about, well, there are about 170 pathways.

 

  4             [Slide.]

 

  5             So, if we look at the response in terms of

 

  6   pathways, we can begin to say, well, are there

 

  7   pathways that are up or down-regulated, and,

 

  8   indeed, this is an example in the case of the

 

  9   experiments I showed you a moment ago.

 

 10             What you see on the lefthand side is the

 

 11   regulon associated with the peroxide response for

 

 12   E. coli to high oxygen--excuse me--on the lefthand

 

 13   side to the superoxide response, the righthand side

 

 14   is the peroxide response.

 

 15             What you can see by the elevated levels of

 

 16   the genes in the superoxide response, that E. coli

 

 17   reacts with the operon, superoxide dismutase and

 

 18   some other enzymes, and the peroxide response is

 

 19   transient, if anything at all.

 

 20             This tells us where the problem is.  The

 

 21   problem is associated with the small amount of

 

 22   superoxide radical that is being made.

 

 23             [Slide.]

 

 24             When we look at clusters of genes, one can

 

 25   see that the green ones are up-regulated in the

 

                                                               243

 

  1   presence of oxygen, and when they are red, they are

 

  2   down-regulated, which happens in the case of

 

  3   nitrogen, and you see green dots with superoxide,

 

  4   but one of the other strange things is that you see

 

  5   proteins that have iron/sulfur in them

 

  6   up-regulated.

 

  7             Why would any self-respecting E. coli

 

  8   up-regulate genes associated with iron/sulfur

 

  9   proteins when you are making a recombinant protein?

 

 10   This didn't make sense, and, in fact, it has

 

 11   nothing to do with the production of alpha-1

 

 12   antitrypsin, but rather has to do with the fact

 

 13   that a small amount of superoxide, that free

 

 14   radical, knocks out the iron/sulfur clusters.

 

 15             There are about 100 proteins in E. coli

 

 16   that have them.  Those proteins are not functional,

 

 17   so how does the cell respond?  It up-regulates

 

 18   pathways in order to compensate.

 

 19             [Slide.]

 

 20             So, within these global techniques, you

 

 21   can begin to understand where the problem is and

 

 22   think about the strategies to better design the

 

 23   process, and basically, it is about taking the next

 

 24   step.

 

 25             Where is the appropriate next step?  I

 

                                                               244

 

  1   will leave it to your imagination whose feet they

 

  2   are.

 

  3             [Slide.]

 

  4             A little quick self-assessment.  When we

 

  5   introduce a process to make a biotherapeutic

 

  6   product, do we know the optimum conditions for

 

  7   quality and quantity of the product today?

 

  8             No, and as a consequence, once the process

 

  9   is in place, we see very substantial process and

 

 10   product improvement during the course of operation,

 

 11   and that is good because it means that we have

 

 12   recognized that there is going to be variance and

 

 13   that we have recognized that we can manage that

 

 14   variance, we can learn from it, and collectively

 

 15   benefit.  That is the reality.

 

 16             So, there are lessons learned there.  The

 

 17   variance that is going to occur is not something to

 

 18   be avoided, it is something to embrace and learn

 

 19   how to manage, and it is getting the right balance

 

 20   of managing that risk.

 

 21             So, during routine manufacturing, do we

 

 22   improve the product in the process?  Absolutely.

 

 23             [Slide.]

 

 24             What is the way forward?  Well, is there a

 

 25   better way than incremental adjustments to optimize

 

                                                               245

 

  1   and scale a process?  Sure, and I think the idea of

 

  2   taking these complex processes and learning how to

 

  3   operate large numbers to capture design of

 

  4   experiments and to capture what happens in that

 

  5   space, and learn how to assess the

 

  6   interdependencies of the parameters is a very

 

  7   exciting opportunity.

 

  8             The technologies that allow us to do it,

 

  9   both from a process side, from an analytical side,

 

 10   from a data analysis side are really important to

 

 11   bring together, and we are not there, but we can be

 

 12   there.

 

 13             We need to live with variance and take an

 

 14   adequate opportunity to learn from that variance.

 

 15   Listen to the data, don't ignore it, listen to it.

 

 16             In doing that, we can again grasp much

 

 17   more experimental space both in variables, as well

 

 18   as time.  So, this issue of embracing that

 

 19   variance, learning what it is about, learning where

 

 20   the problem is, and then using that to come back

 

 21   and develop a robust process, this is the kind of

 

 22   mind-set that PAT is about, and biological

 

 23   processes are very much in need of being thought

 

 24   about and treated and respected in this way.

 

 25             [Slide.]

 

                                                               246

 

  1             In closing, the last slide is to look at

 

  2   what are some of the process evaluation tools.

 

  3   This is not all-inclusive, but it is meant to

 

  4   reinforce just a couple of points that I have made.

 

  5             One is the leverage analytical technology

 

  6   on process and products, what does this really

 

  7   mean?  This is PAT, and this is leading us to a

 

  8   process understanding and a process evaluation.

 

  9             That is very much what it means, that we

 

 10   need to be able to look at the process globally,

 

 11   and not just locally.  It fits exactly in with the

 

 12   guidance that has been laid out for PAT.

 

 13             We need to explore the biological space

 

 14   and the parameter variance.  We need to understand

 

 15   how this variance propagates through a process.

 

 16             It is very interesting, if you take

 

 17   process simulation tools, and we can do a very nice

 

 18   process simulation on any of these processes, and

 

 19   then you do things like Monte Carlo simulation

 

 20   where you have variance in the process, you can

 

 21   begin to understand how that variability at

 

 22   multiple steps is going to propagate through very

 

 23   complex processes.

 

 24             As a consequence, when you do that, you

 

 25   then are not surprised by how a little bit of

 

                                                               247

 

  1   variance here, a little bit of variance there,

 

  2   propagates to give you what the end result is going

 

  3   to look like.  So, with simulation and these tools,

 

  4   you can avoid some surprises.

 

  5             We need to better interrogate the cell at

 

  6   the molecular scale, and then be able to do the

 

  7   multi-scale analysis to scale up.  So, part of what

 

  8   I think PAT is about, is multi-scale analysis,

 

  9   driving down to understand the science, so we can

 

 10   understand where the problem is, and then driving

 

 11   back up with appropriate solutions to eliminate the

 

 12   right problem, in the right way, at the right time.

 

 13             A lot of this about understanding these

 

 14   interdependencies in what is a very large

 

 15   experimental space.

 

 16             Lastly, understanding this connection

 

 17   between the molecular processes, process

 

 18   performance, and product quality.  We are doing I

 

 19   think an exciting job with drug substances in this

 

 20   regard, and we are perfectly capable of carrying

 

 21   that over, with work, to biological products, as

 

 22   well.

 

 23             I will stop there and I hope that I have

 

 24   generated more questions than providing answers,

 

 25   because that is what I started out to do.

 

                                                               248

 

  1             DR. KIBBE:  Thank you.  If there anybody

 

  2   who has any quick questions you want to take care

 

  3   of now before I go on, any point of understanding?

 

  4             [No response.]

 

  5             DR. KIBBE:  In that case, Dr. Koch.

 

  6             DR. KOCH:  I have had the benefit today of

 

  7   a number of speakers who were leading up to the

 

  8   type of things that I wanted to say.  I left out

 

  9   some things, and those of you who have paged

 

 10   through the slides probably can't believe that.

 

 11   There is a lot of slides there, it is going to be a

 

 12   little bit like a fire hose here for a while.  I am

 

 13   going to try to stick to things that are more of a

 

 14   miniature nature or micro-analytical rather than

 

 15   hitting the broad base of all analytical.

 

 16             Let me move into it and I think I will tie

 

 17   in with some of the previous speakers.

 

 18             [Slide.]

 

 19             PAT.  We have heard a number of

 

 20   definitions of it, but again it is looking at all

 

 21   aspects from the chemistry tools through the

 

 22   control strategies and into the data handling

 

 23   aspects.  The goal again, process understanding.

 

 24             [Slide.]

 

 25             The origin of PAT goes back, oh, 50 years

 

                                                               249

 

  1   at least, and we have got a few examples that go

 

  2   back to the mid-forties with some of the German

 

  3   chemical companies applying it, so it is not as if

 

  4   the approach is new.

 

  5             We can go into all of the reasons why it

 

  6   is relatively new in the pharma industry, but that

 

  7   is mostly psychological.  It started within the

 

  8   analytical chemistry labs where tools used for

 

  9   specifications, et cetera, as coming from the areas

 

 10   listed here, were then made portable for running in

 

 11   the process or close to where the process was, and

 

 12   adopting the term "real time analysis."

 

 13             [Slide.]

 

 14             That real-time data resulted in a number

 

 15   of things, in fact, almost every time one went into

 

 16   a process, and this is borrowing from the

 

 17   petrochemical experience, almost every time a

 

 18   sample was taken to a chemical analysis lab, we

 

 19   found out that the results were different if we did

 

 20   it in real time, taking and watching things that

 

 21   you could see fleeting intermediates or a number of

 

 22   things that were indicating both safety and

 

 23   environmental problems.

 

 24             It also was a very good scoping tool for

 

 25   understanding what type of issues and what places

 

                                                               250

 

  1   in a process could be monitored, process

 

  2   understanding results from doing this.

 

  3             [Slide.]

 

  4             What is appropriate for PAT?  It really

 

  5   comes down to a very broad statement, and that

 

  6   anything that gives you data that you are presently

 

  7   not measuring, certainly want to look at cheaper

 

  8   and more reliable, and then we are entering into

 

  9   something here where we are going to get more data

 

 10   than we ever wanted, but we are going to want

 

 11   additional data points in order to build better

 

 12   models from which to control from.

 

 13             This is probably going to be the crack in

 

 14   the wall for Bayesian type approaches where you

 

 15   have to make assumptions because you finally get

 

 16   too much data that you can't possibly study all of

 

 17   it.

 

 18             It is also going to allow us to depart

 

 19   from traditional analytical science technologies,

 

 20   that list that showed up before as coming out of

 

 21   the analytical laboratories, have to move away from

 

 22   that.

 

 23             [Slide.]

 

 24             We are going to have to look at fully

 

 25   integrated analyzer systems.  Historically,

 

                                                               251

 

  1   analysis is detection.  The thing that people have

 

  2   avoided forever is the problem with sampling, I

 

  3   think taking inadequate representative sample to be

 

  4   analyzed.

 

  5             Then, the other thing that often was

 

  6   slipped over because of expense and capability had

 

  7   to do with collecting the data and making sense out

 

  8   of it, and eventual information and knowledge.

 

  9   That has to be all integrated into a system.

 

 10             The next point has to do with inferential

 

 11   analysis, and we have heard that referred to a

 

 12   couple of times, and that is where you can project

 

 13   to the desired product properties by doing some

 

 14   measurement during the process, and it doesn't have

 

 15   to be the property itself, but you have enough data

 

 16   that you can extrapolate to that point.

 

 17             [Slide.]

 

 18             Then, you have to revisit some of these

 

 19   underutilized, but not revolutionary techniques.

 

 20   The few that I mention here are technologies that

 

 21   were discovered in the early 1900s, but not used

 

 22   forever, largely because of instability of optics

 

 23   or computer possibilities back when it was first

 

 24   looked at.

 

 25             [Slide.]

 

                                                               252

 

  1             I want to mention a couple of these.  One

 

  2   is in the optical low coherence reflectometry, that

 

  3   when you do that type of measurement, the result

 

  4   you get depends on things like on the column on the

 

  5   left, the thickness, the particle size,

 

  6   concentration, shape, and some of these other

 

  7   morphological things all affect the measurement.

 

  8             As a result, if you can interpret the

 

  9   signal that you get from the measurement, you can

 

 10   then use it to monitor a number of things.  There

 

 11   are examples there, that are largely from a

 

 12   chemical and materials point of view, but

 

 13   eventually, you get down to being able to monitor

 

 14   tablet coating.

 

 15             The technique started in measuring coating

 

 16   of airplane wings, and we found that that could be

 

 17   extrapolated quickly to other measurements that is

 

 18   being used now for tablet coating, as I mentioned,

 

 19   and we are finding that there is variations during

 

 20   a fermentation or a biological process that can be

 

 21   monitored, and it is a technique that operates at

 

 22   high concentration, in slurries of 70 to 80 percent

 

 23   as a technique for particle size versus the

 

 24   historical need for dilution.

 

 25             [Slide.]

 

                                                               253

 

  1             A couple of examples.  You can look at a

 

  2   multi-layer film.  Here is an example of a drug

 

  3   delivery patch.  I think you can see some of the

 

  4   peaks there on the bottom.

 

  5             Very interestingly, what happens in this

 

  6   process, it looks like a chromatogram with various

 

  7   peaks, however, it is the bounce back of the

 

  8   photons at each layer, and you measure the time

 

  9   that it takes to come back and project into

 

 10   distance.

 

 11             Each one of those peaks is a layer.  It is

 

 12   a layer from the barrier layer on the outside and

 

 13   the back, and then the intermediate layer is

 

 14   between active ingredients, so it becomes a way to

 

 15   measure how much active ingredient one has placed,

 

 16   so the baseline is basically the thickness of the

 

 17   active ingredient.

 

 18             The scattered material example is one

 

 19   where you have a total reflection of the photon and

 

 20   the path in which it travels indicates the

 

 21   complexity of the mixture, and you can extrapolate

 

 22   then into things like particle size, shape, and

 

 23   waveguide formation, et cetera.

 

 24             [Slide.]

 

 25             An example of being able to look at

 

                                                               254

 

  1   consistency, there is one curve here that shows at

 

  2   one concentration, you can see quite a range of

 

  3   small particles from basically 20 to 90 nanometers,

 

  4   or you can take one size, in the lower example, of

 

  5   308 nanometers, and get a concentration difference.

 

  6   So, it has proven to be quite valuable in that

 

  7   regard.

 

  8             [Slide.]

 

  9             Moving on to Raman, certainly, everyone

 

 10   has heard the terminology, but as you look at some

 

 11   of the potential advantages now that the stability

 

 12   of the lasers have improved in some of the data

 

 13   handling, and as databases grow, you can look at

 

 14   non-invasive or non-destructive technology.

 

 15             You can work in aqueous systems.  You can

 

 16   do multiplex of your instrument using fiber optics

 

 17   that can go hundreds of meters, and you can also

 

 18   then look at chemical structure and fingerprinting

 

 19   of both inorganic and organic materials.

 

 20             [Slide.]

 

 21             Then, with effective probes, in fact, this

 

 22   particular probe that is demonstrated here, was the

 

 23   one that we used in the practicum and moved between

 

 24   the various centers to study some milling and

 

 25   mixing operations, but we have done a number of

 

                                                               255

 

  1   things in composition, as well as, at the bottom

 

  2   right, putting it in a protein mixture in terms of

 

  3   determining aspects of that material.

 

  4             [Slide.]

 

  5             The fringing electric field or

 

  6   dielectrometry sensor is pretty simple, one that

 

  7   was developed for detecting mines, and it has to do

 

  8   with the ability to set your electrical fields with

 

  9   the various sensors in setting the distance and the

 

 10   intensity, and you can get a disturbance of that

 

 11   electrical field based on the properties of the

 

 12   sample.

 

 13             You can measure things like density,

 

 14   distance from the sensor, texture, and moisture,

 

 15   and moisture not only in concentration, but

 

 16   distribution, so you will start to look at filter

 

 17   cakes or other aspect of various processes.  You

 

 18   have another relatively unused method that can be

 

 19   applied.

 

 20             [Slide.]

 

 21             To date, a number of things happening in

 

 22   the paper pulp industry, pharmaceutical products,

 

 23   and we have got a few companies, pharma-based, that

 

 24   are using it for mixing consistency, a lot of food

 

 25   applications including some of the baking companies

 

                                                               256

 

  1   to monitor the moisture distribution in cookies and

 

  2   cakes and things, and that turns out to be pretty

 

  3   important for them, composites, plastics, et

 

  4   cetera.

 

  5             [Slide.]

 

  6             Going on to surface plasmon resonance, a

 

  7   number of things, primarily in miniaturization and

 

  8   sensitivity have occurred here, and plugging some

 

  9   disciplines together from electrical engineering

 

 10   and genetic, have come up with some real-time

 

 11   biosensors that are operating at a very fast mode.

 

 12             [Slide.]

 

 13             Work sponsored by the Department of

 

 14   Defense, again that tie in with some of the things

 

 15   we heard earlier on homeland security.

 

 16             You can start to look at high throughput

 

 17   screening, automated protein purification, and

 

 18   number of toxins, food-related activities, and we

 

 19   are actually moving quite rapidly into response in

 

 20   the food industry for safety, security, nutrition

 

 21   in the food and related water chains.

 

 22             [Slide.]

 

 23             One example, this has been demonstrated in

 

 24   a protein purification system, would be a way in

 

 25   which after the broth is separated and some

 

                                                               257

 

  1   chromatography applied using biosensors, one can

 

  2   determine when to change columns or monitor the

 

  3   process, which brings us to biosensors and the need

 

  4   in the bioprocess in general.

 

  5             [Slide.]

 

  6             I have been in discussions with Harry Lam

 

  7   of Genentech, to get a feel for what type of things

 

  8   the industry is looking at, and certainly to

 

  9   maintain a consistent product performance or

 

 10   process performance with the development cycle from

 

 11   early stage through manufacturing.

 

 12             [Slide.]

 

 13             Measurement is needed in order to look at

 

 14   the underlying functional relationships that occur

 

 15   in the process, as well as some of these

 

 16   interactions of the organisms with their

 

 17   environments.

 

 18             [Slide.]

 

 19             We need to improve the capabilities for

 

 20   process control, and the type of measurements are

 

 21   going to be broad based, biological, chemical,

 

 22   physical.

 

 23             [Slide.]

 

 24             Much of this has been touched on today -

 

 25   biological with this whole range of things that are

 

                                                               258

 

  1   of a cellular nature.

 

  2             [Slide.]

 

  3             Chemical, we have got a number of things

 

  4   in the media that need to be addressed, that have

 

  5   to do with the nutrients and the additives, et

 

  6   cetera.

 

  7             [Slide.]

 

  8             It continues on when you start to

 

  9   characterize the product, the by-products, the

 

 10   environment, as well as the off-gas.

 

 11             [Slide.]

 

 12             Physical.  We have heard much of this in

 

 13   terms of the type of things that need to be looked

 

 14   at.

 

 15             [Slide.]

 

 16             What can we look at today?  Much of this

 

 17   was mentioned here in the last couple of

 

 18   presentations, of things that are being used to

 

 19   monitor, but that leaves a number of the issues on

 

 20   the table yet to be addressed and solved.

 

 21             [Slide.]

 

 22             Also, the industry is looking at the

 

 23   various requirements that are going to be

 

 24   necessary, and it is a lot more than just having a

 

 25   measurement tool, but to get into the things that

 

                                                               259

 

  1   have to do with sterilization, interference, and

 

  2   the fouling, low maintenance, and the small size.

 

  3             We have heard several assumptions today

 

  4   that if it's smaller, it could be better.

 

  5             [Slide.]

 

  6             That gets us into what has been driving

 

  7   the improvements in measurement over the last, say,

 

  8   20 years, and it has been the advances in

 

  9   miniaturization.  Much of this has been driven by

 

 10   technologies in the computing industry and the

 

 11   ability to make things smaller and use microfluidic

 

 12   technologies, et cetera.

 

 13             Certainly, new materials, the optic

 

 14   advances, and computing have helped, but

 

 15   miniaturization is really a big one.

 

 16             [Slide.]

 

 17             It has been focus of the center where I am

 

 18   located in Washington.  It has been a

 

 19   multi-industry, and I have implied that a few

 

 20   times.  A number of industry come together and

 

 21   discuss advance in real-time measurement, and we

 

 22   are now beginning to apply those things to the

 

 23   food, pharma, biotech industry.

 

 24             [Slide.]

 

 25             Multidisciplinary.  There are many

 

                                                               260

 

  1   examples where bringing different disciplines

 

  2   together results in some very interesting sparks

 

  3   coming from that smoke, presently supporting 20

 

  4   different research projects at 5 universities, and

 

  5   involved with some international collaboration.

 

  6             [Slide.]

 

  7             The initiatives, and we will see the

 

  8   importance of this growing, is sampling and

 

  9   sensors.  That is one that we try to act as a forum

 

 10   across industry.  Trying to also compile analytical

 

 11   and chemometric methods, what to use in terms of

 

 12   interpreting the data.

 

 13             A couple of things that are used just

 

 14   inside for the members, are to look at

 

 15   micro-instrumentation for the high throughput

 

 16   experimentation, the CombiChem, and some of the

 

 17   process optimization tools, and then a fermentation

 

 18   platform, and I will mention some of the things

 

 19   there.

 

 20             [Slide.]

 

 21             When we look at this response to high

 

 22   throughput experimentation, we get into the

 

 23   micro-instrumentation world, but also the

 

 24   micro-reactor world.  I have to agree with that

 

 25   Charles mentioned, the petrochemical industry is

 

                                                               261

 

  1   finding huge benefits in scaling down before you

 

  2   scale up, and going down to molecular interactions

 

  3   in a number of data-gathering aspects at the small

 

  4   scale to understand how to then move on from that

 

  5   to macro scales.

 

  6             [Slide.]

 

  7             We also have a number of techniques that

 

  8   are being miniaturized largely due to advances.  As

 

  9   I mentioned before, most of the analytical

 

 10   technologies, we only have a few that have not been

 

 11   miniaturized yet or taken on-line, and some of

 

 12   those are microscopy-based, but we actually have

 

 13   some breakthroughs now in bifringes and other

 

 14   things that could help in this respect.

 

 15             [Slide.]

 

 16             I will give you a couple of examples.  In

 

 17   micro-LC, we have got a small 100-micron flow

 

 18   channel where you mix a sample at a mobile phase

 

 19   and then detect the deflection in your laser beam

 

 20   with a position-sensitive detector.

 

 21             [Slide.]

 

 22             We have since found, after starting into

 

 23   this project, that low molecular weight material

 

 24   diffuses much faster than the higher molecular

 

 25   weight material.

 

                                                               262

 

  1             [Slide.]

 

  2             So, why not put two sensors in-line and

 

  3   then begin to calculate the difference between

 

  4   those distances in terms of a particular molecule.

 

  5             [Slide.]

 

  6             What has resulted is an in-line molecular

 

  7   mass sensor where we are able, in this case, to

 

  8   look at polyethylene glycols from a very low

 

  9   molecular weight. Actually, it has now been taken

 

 10   to over 100,000 molecular weight in terms of a

 

 11   standard curve.

 

 12             [Slide.]

 

 13             This has resulted in other things now, in

 

 14   some biological testing where we can see peptide

 

 15   synthesis, we can look at polysaccharide synthesis

 

 16   and be able to see differences as chains are

 

 17   building, and also be able to see differences in

 

 18   diffusion in following trends in that way.

 

 19             [Slide.]

 

 20             Developments at Sandia, again, homeland

 

 21   security basis, have resulted in a micro chem lab.

 

 22   This is a very interesting thing, obviously, the

 

 23   size of a dime is quite impressive, but when you

 

 24   look at the SAW ray detector you have go a 1-meter

 

 25   column, and your sample pre-absorption.

 

                                                               263

 

  1             [Slide.]

 

  2             You put all that into a hand-held unit,

 

  3   this is a now a hand-held GC, but the end there

 

  4   indicates it is also an LC, so that has all been

 

  5   incorporated into taking today's lab technology

 

  6   down to a very small size.

 

  7             [Slide.]

 

  8             Some work that we have been involved with

 

  9   recently is when you go to use of nanoparticles in

 

 10   your column, you can increase the speed.  We are

 

 11   now talking of these compounds being separated in

 

 12   two seconds.

 

 13             Normally, you are looking at 40-minute

 

 14   type turnarounds on a lot of these GC analysis

 

 15   things that have been improving, and I can't really

 

 16   talk about it, but we now have a similar separation

 

 17   in 500 milliseconds, that things are really flying

 

 18   in that way, so it has become a real-time

 

 19   analytical technology.

 

 20             [Slide.]

 

 21             A small mass spec has been developed.

 

 22   There is three or four examples of taking mass spec

 

 23   down to these small sizes.

 

 24             [Slide.]

 

 25             We are also involved with development at

 

                                                               264

 

  1   UC/Davis with the micro labs, the electrical and

 

  2   computing and the food science areas, to develop a

 

  3   NMR.

 

  4             [Slide.]

 

  5             This is an NMR now that early signal is

 

  6   shown on the bottom left, which showed water, a lot

 

  7   of excitement by the food group because they could

 

  8   monitor a number of things in real time. It has

 

  9   since been refined to the bottom right there, and

 

 10   it has been taken from a protein signal, we have

 

 11   now seen carbon and phosphorus, so we are talking

 

 12   about a hand-held NMR that is going to be

 

 13   multinuclear and have a cost of probably under

 

 14   $20,000.

 

 15             [Slide.]

 

 16             So, all these advances in sensors and

 

 17   controls again highlight the need, how do you get

 

 18   the right sample to these technologies.

 

 19             [Slide.]

 

 20             The chemical industry has come to us, and

 

 21   we have been a forum for discussions on how to

 

 22   create new sampling and standardized technologies

 

 23   in that arena.

 

 24             [Slide.]

 

 25             The typical sampling in a petrochemical

 

                                                               265

 

  1   plant is a large, often covering a wall, quarter of

 

  2   a million dollars worth of instrumentation just to

 

  3   interface the process with the analyzer.

 

  4             [Slide.]

 

  5             That has now shrunk down to an inch and a

 

  6   half by an inch and a half modules, a standard set

 

  7   by the ISA, and this platform now houses the valves

 

  8   and filters and regulators to interface again the

 

  9   process with the analyzer.

 

 10             [Slide.]

 

 11             What has been evolving here, this concept

 

 12   started in late 2000, and it has now generated to

 

 13   point where we are beginning to think of how we

 

 14   could make this Smart and how to utilize advances

 

 15   in micro-analytical.

 

 16             [Slide.]

 

 17             So, the base here has been defined.  We

 

 18   now have a standard sampling interface that can be

 

 19   heated or cooled, or whatever, and the flow

 

 20   patterns all defined, and in the next couple of

 

 21   months, we are standardizing a connectivity.

 

 22             This is getting into some control

 

 23   engineering terminology of how do you move the

 

 24   signals from that platform to distributor control

 

 25   systems and other fields of how do you use that.

 

                                                               266

 

  1             [Slide.]

 

  2             Then, what has happened is you can now

 

  3   drop your pressure regulators, your valves, and

 

  4   your filter onto that platform and be able to

 

  5   monitor what they are doing.

 

  6             A very interesting story happened at again

 

  7   interfacing the process with the analyzer.  The

 

  8   first year of use of these devices caused the

 

  9   engineers to say why does the analyzer have to be a

 

 10   refrigerator size, when the sampling system has

 

 11   come off the wall to this fairly small

 

 12   compartmentalized unit.

 

 13             So, this platform has now become the base

 

 14   for micro-analytical, so it has become a standard

 

 15   platform for the development of micro devices.

 

 16             Three or four years ago, if somebody had

 

 17   come in with a small GC and say wow, isn't this

 

 18   neat, and we would say that is really nice, but how

 

 19   do we use it, how do we go to this big,

 

 20   wall-mounted sampling system and put this little GC

 

 21   at the end of it.

 

 22             That has changed, people are now putting

 

 23   on a fair amount of suction for the development of

 

 24   these devices.

 

 25             [Slide.]

 

                                                               267

 

  1             So, we predict, and it is beginning to

 

  2   happen, that the NeSSI platform will become the

 

  3   base for a micro-analytical lab.  Already we have

 

  4   oxygen and pH and moisture, mass flow controllers,

 

  5   little mass specs, all of the techniques that are

 

  6   listed there have the plan to be mounted on this

 

  7   particular platform.

 

  8             [Slide.]

 

  9             And then we have been devising different

 

 10   interfaces.  Our Raman sensor now will fit on the

 

 11   NeSSI platform.

 

 12             [Slide.]

 

 13             The surface plasmon resonance, this is the

 

 14   one that does the very fast biological detection,

 

 15   is now down to the size where the flow channels

 

 16   will interface with the surface and provide almost

 

 17   real-time biological detection in the NeSSI

 

 18   platform.

 

 19             [Slide.]

 

 20             And we have taken something that basically

 

 21   used to be flow injection analysis, it migrated to

 

 22   be called sequential injection analysis, now it is

 

 23   micro sequential injection analysis, where you can

 

 24   put wet chemistry on a multi-functional,

 

 25   multi-position valve, so you can scale down wet

 

                                                               268

 

  1   chemistry and titrations and things, and do things

 

  2   like glucose, nitrogen, nutrients, and inorganic

 

  3   detection, and this is now on the NeSSI platform.

 

  4             [Slide.]

 

  5             So, there is almost nothing right now that

 

  6   we don't have that couldn't possibly fit on here,

 

  7   and we see it, not only for the process control,

 

  8   but all kinds of optimization studies that could

 

  9   interface with lab-based fermentation and with the

 

 10   micro-reactor systems for the chemical world.

 

 11             [Slide.]

 

 12             The last thing I will mention is our

 

 13   Fermentation Initiative.

 

 14             [Slide.]

 

 15             We are trying to apply the known

 

 16   techniques and compare them with things that are

 

 17   evolving and have applications in other fields.  We

 

 18   want to provide training and understanding the

 

 19   implication of some of these measurements.

 

 20             [Slide.]

 

 21             We have set up some platforms that are now

 

 22   outfitted with this array of instrumentation, and

 

 23   you can see things like dielectric spectroscopy,

 

 24   the surface tension, light reflective spectroscopy,

 

 25   et cetera, that are not traditionally being used in

 

                                                               269

 

  1   the fermentations, but we are gathering data and

 

  2   finding then ways to extrapolate to which

 

  3   fermentation areas they will best influence, and

 

  4   then looking at sampling.

 

  5             Sampling and fermentation is a big

 

  6   problem.  Go back to some of the things that we put

 

  7   together that Genentech summarized.

 

  8             [Slide.]

 

  9             They are very concerned about how to

 

 10   achieve these type of considerations, and then you

 

 11   get into some of the sterile requirements and how

 

 12   do you design your sampling system, so it will meet

 

 13   these requirements.

 

 14             [Slide.]

 

 15             So, what we have is a plan to continue to

 

 16   scope out activities from an analytical point of

 

 17   view, but to implement and evaluate this NeSSI

 

 18   platform for not only sampling, but sensor and

 

 19   process control interfaces, so we have a platform

 

 20   now that is being put in to work with sampling the

 

 21   broth and another one with head space.

 

 22             We are looking at chemometric tools to

 

 23   model this batch variability and look at various

 

 24   data fusion approaches.  We need to do this to

 

 25   begin to develop these automated tools to evaluate

 

                                                               270

 

  1   production data and implement chemometrics as much

 

  2   as possible for quantifying process performance and

 

  3   applying these PCA approaches to performing

 

  4   automated pattern recognition.

 

  5             [Slide.]

 

  6             To borrow a little bit from Helen's

 

  7   earlier slide, it is going to be an exciting time.

 

  8   We have got a lot of things in front of us, but to

 

  9   take the advances in PAT from the other industries,

 

 10   through the pharmaceutical on to the biological, I

 

 11   think is going to be very rewarding.

 

 12             DR. KIBBE:  Thank you.

 

 13             Are there any quick questions?

 

 14             DR. SINGPURWALLA:  I have a comment.

 

 15             You have this nice chart on

 

 16   multidisciplinary, page 16, and you also had CPAC

 

 17   initiatives.  Just from a parochial point of view,

 

 18   I noticed the absence of a statistician, yet, you

 

 19   are discussing the sampling, which is really a

 

 20   statistical issue.

 

 21             DR. KOCH:  You are right.  In fact, that

 

 22   list is not complete.  What that list is, is the

 

 23   present principal investigators involved with our

 

 24   programs.  We have just finished a project with the

 

 25   chairman of our Statistics Department where we were

 

                                                               271

 

  1   funding things just for what you are saying.

 

  2             So, really, that list is project

 

  3   dependent.  I could probably add as many as six

 

  4   other areas, like physics was on there, and a few

 

  5   others in the past year, so we rotate projects in

 

  6   and out.  Chemometrics is probably based in

 

  7   statistics.  They don't like to admit it.

 

  8             DR. KIBBE:  Anything else?  Ajaz doesn't

 

  9   want to comment?  Okay.

 

 10             Tom, wrap us up.

 

 11             DR. LAYLOFF:  Much of what I wanted to say

 

 12   has been said already, so I will speed through my

 

 13   slides, and I have a few comments at the end that

 

 14   are not in the slides.

 

 15             [Slide.]

 

 16             First of all, PAT, with the subcommittee

 

 17   to this committee, advisory committee, we had a

 

 18   series of charges which were given to us.

 

 19             [Slide.]

 

 20             We had meetings lasting through 2002,

 

 21   three meetings.  We covered applications and

 

 22   benefits, process and analytical validation,

 

 23   chemometrics, process-product development, process

 

 24   and analytical validation, a proposed PAT training

 

 25   and certification program, which I think was one of

 

                                                               272

 

  1   the highlights of the activities.

 

  2             Computer systems validation, 21 CFR 11,

 

  3   and Joe out there tackled that one, PAT case

 

  4   studies, and rapid microbiological testing was

 

  5   tacked on near the end there.

 

  6             [Slide.]

 

  7             We reported that back to this committee

 

  8   back in October.  There was a definition of process

 

  9   analytical technology.  I am not going to read that

 

 10   to you again, you have already seen it.

 

 11             [Slide.]

 

 12             Again, more statements on PAT

 

 13   applications.

 

 14             [Slide.]

 

 15             Now, this was not included.  Historically,

 

 16   there has never been anything to stop people from

 

 17   using new technologies.  As a matter of fact, in

 

 18   the 1978 preamble to the CGMPs, there is no

 

 19   prohibition in the regulations against the

 

 20   manufacturing of drug products using better, more

 

 21   efficient, and innovative methods.

 

 22             It is a big box, and it has been there

 

 23   since 1978.

 

 24             The USP also allows alternative methods

 

 25   for assessments.

 

                                                               273

 

  1             [Slide.]

 

  2             The committee proceeded by coming up with

 

  3   a general guidance through Raj Uppoor and the OPS

 

  4   staff to generate a guidance, which defined a

 

  5   regulatory position for the process and added some

 

  6   incentives.  Then, the FDA PAT team, which came

 

  7   through the training program you have heard about

 

  8   earlier.

 

  9             [Slide.]

 

 10             Of course, the Agency's perspectives.  One

 

 11   of the things that I think is very interesting,

 

 12   coming from many years of service in the Agency,

 

 13   was the Agency's use of existing knowledge,

 

 14   experience, and guidances from other FDA

 

 15   components, and NIST, ASTM, and ANSI.

 

 16             The FDA tended over the period of my

 

 17   tenure to be very introspective, if it wasn't NIH,

 

 18   not invented here in FDA, we had very little use

 

 19   for it.  Going to ASTM and NIST was more of an

 

 20   engineering approach, and we tended to hang with

 

 21   the pharmacists in the USP.

 

 22             The USP, of course, was established by

 

 23   practitioners as a book of recipes to assure

 

 24   quality, and we hung with that, with the

 

 25   practitioners rather than with the engineering.

 

                                                               274

 

  1             ASTM was established by engineers and

 

  2   chemists to deal with defective rails in the

 

  3   railroad, so they tended to be very

 

  4   engineering-oriented, and it was quite interesting

 

  5   that when FDA-CDRH went out looking for standards,

 

  6   they went to the engineering standard type area

 

  7   rather than practitioners of pharmaceuticals.

 

  8             Now, the switch in OPS of looking at ASTM

 

  9   standards is very interesting because it moves

 

 10   process analytical technologies into an arena where

 

 11   there are engineers and chemists, scientists rather

 

 12   than practitioners.  It's a switch in philosophy.

 

 13             Also, the ANSI and ISO fit in that also.

 

 14   They established a framework for manufacturers with

 

 15   flexibility needed to develop new designs.

 

 16             [Slide.]

 

 17             Future issues.  Validation data and

 

 18   retention.  We have heard some about retention of

 

 19   data, and I don't think that has been addressed

 

 20   well, but the process analytical technology is

 

 21   going to deluge with information, and there is

 

 22   going to have to be some way of defining what is

 

 23   essential and should be retained, and what is not

 

 24   essential.

 

 25             The definition of in-process endpoint

 

                                                               275

 

  1   detection, data acquisition and storage.  In a

 

  2   process of PAT, you have to have some component in

 

  3   the process which is measurable and defines an

 

  4   endpoint.  You have to have analytics, but you have

 

  5   to have something that you are looking for.

 

  6             The documentation of the data acquired and

 

  7   electronic signature closures of decision points

 

  8   are going to be an issue, and the incoming material

 

  9   stream consistency and robustness assessments are

 

 10   going to be critical for supporting PAT also.

 

 11             [Slide.]

 

 12             Regulatory incentives, we have gone over

 

 13   those already, not a requirement.

 

 14             [Slide.]

 

 15             How to move forward, try and do it by

 

 16   evolution rather than revolution.  Don't bring it

 

 17   all up at once.

 

 18             [Slide.]

 

 19             And the guidance which came out in

 

 20   September, just a few items from it.

 

 21             [Slide.]

 

 22             The guidance is intended to describe a

 

 23   regulatory framework that will encourage the

 

 24   voluntary development and implementation of

 

 25   innovative pharmaceutical manufacturing and quality

 

                                                               276

 

  1   assurance--manufacturing and quality assurance,

 

  2   voluntary, innovative.  Those are key terms.

 

  3             [Slide.]

 

  4             The scientific risk-based framework

 

  5   outline in the guidance should help manufacturers

 

  6   develop and implement new and efficient tools for

 

  7   use during pharmaceutical development,

 

  8   manufacturing, and quality assurance while

 

  9   maintaining or improving the current level of

 

 10   product quality assurance.

 

 11             The framework we have developed has two

 

 12   components:  a set of scientific principles and

 

 13   tools supporting innovation, and a strategy for

 

 14   regulatory implementation that will accommodate

 

 15   innovation-keys.

 

 16             [Slide.]

 

 17             Among other things, the regulatory

 

 18   implementation strategy includes creation of a PAT

 

 19   team approach to the CMC review and CGMP

 

 20   inspections and joint training and certification of

 

 21   PAT review and inspection staff.

 

 22             The Agency is encouraging manufacturers to

 

 23   use the PAT framework described here to develop and

 

 24   implement new pharmaceutical manufacturing and

 

 25   quality assurance technologies.

 

                                                               277

 

  1             [Slide.]

 

  2             The guidance is written for a broad

 

  3   industry audience in different organizational units

 

  4   and scientific disciplines.

 

  5             To a large extent, the guidance discusses

 

  6   principles with the goal of highlighting

 

  7   technological opportunities and developing

 

  8   regulatory processes that encourage innovation.

 

  9             [Slide.]

 

 10             Biologics and PAT.  The umbrella guidance

 

 11   covers biological production within the scope.

 

 12   Presentations before our committee included

 

 13   individuals that were using, or companies that were

 

 14   using, process analytical technology to monitor

 

 15   fermentation and purification of biological

 

 16   materials, so it fits if you can define those kinds

 

 17   of controls.

 

 18             However, the process differences that

 

 19   occur in biologics may require or likely will

 

 20   require additional skills and an expansion of the

 

 21   training and certification program.

 

 22             So, that PAT concept, the training of

 

 23   reviewers and inspectors will probably need to be

 

 24   expanded with training in biologics type PAT

 

 25   applications.  The standard chemical stuff is not

 

                                                               278

 

  1   going to work, you are going to have to expand it

 

  2   beyond that.

 

  3             But I think the concept that came out of

 

  4   our committee of having a training program and a

 

  5   certification of competencies is very useful for

 

  6   building teams to get around some of the silos that

 

  7   we have in FDA, that have been there, because those

 

  8   silos contribute to poor science and poor

 

  9   regulation, some of them, and that needs to be

 

 10   straightened out, and this is a good attempt at

 

 11   beginning to do that.

 

 12             [Slide.]

 

 13             Acknowledgments.  Ajaz has done a great

 

 14   job, I am a great fan of his efforts in taking this

 

 15   and driving it forward because he has really done a

 

 16   great job of pulling it.

 

 17             Raj for doing the guidance.  My former

 

 18   colleagues at the DPA, DPQR, colleagues that

 

 19   presented at the PAT Committee, and those reports

 

 20   are at that web site.

 

 21             It has been a lot of fun for me to work on

 

 22   the PAT Subcommittee.  As Ajaz said, it was a

 

 23   project that we started about 11 years ago, and to

 

 24   see it come to fruition now has really been great.

 

 25             I think the industry has got to do more,

 

                                                               279

 

  1   they are going to do more as they reduce their

 

  2   inventories and move to just-in-time manufacture,

 

  3   which will reduce cost, and, very importantly, help

 

  4   bring the vision of health to all closer.

 

  5             That's it.  Any questions?

 

  6             DR. KIBBE:  Any questions for Tom?  Now,

 

  7   we understood that you had answers to all of these

 

  8   questions from Keith Webber.

 

  9             DR. LAYLOFF:  I do, I do.

 

 10             DR. KIBBE:  If you could just tell us what

 

 11   the answers are, we could all go to--Happy Hour,

 

 12   right.

 

 13             No questions?  Perhaps, Keith, you would

 

 14   like to lead us through these questions and try to

 

 15   get at least some of our collective wisdom on some

 

 16   of them.

 

 17             Committee Discussion and Recommendations

 

 18             DR. KIBBE:  What technologies are

 

 19   available now to evaluate the characteristics of

 

 20   protein products in real time during manufacturing?

 

 21   Who has an answer?

 

 22             Dr. Koch.

 

 23             DR. KOCH:  I don't know if I could answer

 

 24   that directly, but there are a number of monitoring

 

 25   methods that are being used by those manufacturing

 

                                                               280

 

  1   protein products today, and it probably might be

 

  2   best to pool some kind of a compilation from those

 

  3   who are presently in that product arena.

 

  4             Often from the place where I am sitting, I

 

  5   have a difficult time judging what measurement

 

  6   techniques that we develop, and we have been

 

  7   involved with technologies that have made it into

 

  8   commercialization, but when we ask one of our

 

  9   members is it working or how well is it working,

 

 10   and their processes, we can normally tell just by

 

 11   the smile or lack of it.

 

 12             So, we are in a situation where we are

 

 13   developing tools for a toolbox, and we are never

 

 14   quite sure how well they are being applied.

 

 15             I don't know, Gerry, you are probably in a

 

 16   position, or Rick, I saw earlier.

 

 17             MR. MIGLIACCIO:  My background is on the

 

 18   small molecule side, so I wouldn't want to leap

 

 19   into this.

 

 20             DR. LAYLOFF:  I have a question on what

 

 21   does it mean evaluate the characteristics, because

 

 22   the sequence is pretty well clean on proteins.  Are

 

 23   you talking about secondary, tertiary, quaternary

 

 24   structures?

 

 25             DR. WEBBER:  I am talking about more the

 

                                                               281

 

  1   overall structure, say, tertiary, and in those

 

  2   cases where it is applicable, quaternary

 

  3   structures, but also the post-translational

 

  4   modifications structure of the product, which is

 

  5   one of the I think most variable in terms of what

 

  6   we see to change during, say, fermentation, or it

 

  7   can be selected out during purification.

 

  8             You can get various species of product get

 

  9   selected or rejected during purification, so that

 

 10   is really what I was looking at there.

 

 11             DR. LAYLOFF:  So, it is not a process

 

 12   closure, it is actually an assessment of the

 

 13   product coming out of the process.

 

 14             DR. WEBBER:  In this particular question,

 

 15   yes, it is the product which is not necessarily

 

 16   coming out, but the product during manufacturing.

 

 17   One of the areas that I have seen reported is the

 

 18   ability to use--and I discussed it a little

 

 19   bit--immunological techniques or lectins to look at

 

 20   structures.

 

 21             For example, carbohydrates on products, I

 

 22   haven't seen that in practice yet as a PAT, but

 

 23   that is something that may be coming down the line.

 

 24             DR. KIBBE:  Dr. Cooney.

 

 25             DR. COONEY:  First of all, this is a very

 

                                                               282

 

  1   important question because this is a question that

 

  2   relates what you make to its therapeutic safety and

 

  3   efficacy, on the one hand, so part of addressing

 

  4   this question is to understand the relationship

 

  5   between particularly post-translational

 

  6   modification, glycosylation, acetylation,

 

  7   phosphorylation, and so on, and its therapeutic

 

  8   efficacy.

 

  9             It is also important because a lot of

 

 10   those properties are known to vary with the

 

 11   process, so whatever you use at this point for an

 

 12   assay or an analytical technology, links you back

 

 13   to the process, on one hand, presumably you will

 

 14   understand that linkage, and links you forward to

 

 15   the patient, on the other hand.

 

 16             I think the advances in mass spec that

 

 17   have evolved with proteins and, in particular, with

 

 18   proteins that are modified, is quite substantial.

 

 19   Quite recently, I saw ultra-high pressure

 

 20   chromatography, which takes advantage of a number

 

 21   of innovations in chromatography by being able to

 

 22   go to very small particles with a very high amount

 

 23   of surface area at very high pressures, and by

 

 24   doing that, you can very quickly get a very high

 

 25   resolution of complex mixtures.

 

                                                               283

 

  1             So, a combination of size, shape methods

 

  2   plus mass spectrometry and being able to work with

 

  3   large molecules and very small amounts of material,

 

  4   that seems to be where things are going, and

 

  5   provides a very powerful armamentarium of PATs.

 

  6             DR. KIBBE:  Tom has another comment?

 

  7             DR. LAYLOFF:  I was wondering, on

 

  8   chromatographic procedure, whether or not you could

 

  9   have subsequent post-translational modification of

 

 10   the proteins themselves. Denaturation would be one,

 

 11   but reactions with the supports themselves at

 

 12   15,000 psi are reactive with solvents catalyzed on

 

 13   the supports.

 

 14             I don't know how you validate the

 

 15   separation tools at 15,000 psi.

 

 16             DR. COONEY:  You raise a very good point.

 

 17   The work on ultra-high pressure chromatography is

 

 18   very new and solvents and dissolved gases and

 

 19   solvents are very reactive at those pressures.

 

 20   That needs to be sorted out.  It's the right

 

 21   question, and I think one can design the

 

 22   experiments to get the answer.

 

 23             DR. KIBBE:  I feel like we are doing 1 and

 

 24   2 a little bit.  What tools would allow us to

 

 25   understand the manufacturing process better?  The

 

                                                               284

 

  1   tools that will allow us to understand the product

 

  2   will also allow us to go back and look at the

 

  3   process.

 

  4             Is there anything specifically that

 

  5   anybody would like to add on that?

 

  6             DR. LAYLOFF:  I was going to say one of

 

  7   the things that comes up, of course, in near

 

  8   infrared, in applications, is that you don't have

 

  9   to separate anything, you just look at it, and you

 

 10   define your endpoints on a polyvariate system.

 

 11             It may be also possible to do something

 

 12   like looking at a mass spec fingerprint without

 

 13   separating anything, just look at the mass spec,

 

 14   just hammer it and see what it looks like during

 

 15   the course of a process, just hammer it at

 

 16   intervals and just see what it looks like until you

 

 17   define an endpoint by another source, and then use

 

 18   that as an endpoint indicator.

 

 19             DR. KIBBE:  Anything else you need?

 

 20             DR. WEBBER:  Just one follow-up question

 

 21   with regard to No. 1 from the presentation that Dr.

 

 22   Koch did.

 

 23             You had shown LC and NMR technologies that

 

 24   were miniaturized, and we like to think smaller is

 

 25   better and more PAT-like.  Would those

 

                                                               285

 

  1   technologies, as they are now, be amenable to

 

  2   biotech products or do smaller molecules, would

 

  3   they be useful for looking at, say, fermentation

 

  4   components, and things like that?

 

  5             DR. KOCH:  We plan to have all those

 

  6   techniques tied in with the fermentation project,

 

  7   so there are early reasons to believe that we will

 

  8   be getting data from them.

 

  9             I think one of the important things to

 

 10   point out, when we even talk about DNIR or Raman or

 

 11   some of the others, I think we will find with time

 

 12   an array of technologies with a multivariate

 

 13   evaluation of the data is going to prove in the end

 

 14   to be quite valuable, so that you can look at, and

 

 15   see, the variations that are coming from batch to

 

 16   batch or system to system.

 

 17             DR. WEBBER:  Thank you.

 

 18             We had completed Item No. 2 or not, you

 

 19   sort of led into that, but are there any other

 

 20   comments with regard to what tools would be

 

 21   available to allow us to better understand biotech

 

 22   processes?

 

 23             DR. KOCH:  Maybe just a comment on that

 

 24   one.  It seems like at the top of most

 

 25   manufacturers' list is bioviability, and that takes

 

                                                               286

 

  1   on all kinds of definitions based on what product

 

  2   one is working with.

 

  3             The more tools that are developed to

 

  4   determine the health of the organism, the maturity

 

  5   of the system, or measurement or metrics to

 

  6   determine when is the best time to harvest, there

 

  7   is a number of things I believe are going to be

 

  8   advancing there, both direct and indirect methods.

 

  9             DR. LAYLOFF:  Then, there is also going to

 

 10   probably be indirect methods, like on flowing

 

 11   stream systems, where you actually take the

 

 12   fermentation broth, react it on to other species,

 

 13   which could serve as a surrogate to where the

 

 14   process is located.

 

 15             DR. COONEY:  One of the things that will

 

 16   surely happen in the diagnostics field is improve

 

 17   proteomic techniques.  The genomic techniques are

 

 18   not so bad, the proteomics are still early stage,

 

 19   but as we develop better proteomic techniques, as

 

 20   you develop better immuno-based panels that are

 

 21   important in diagnosis of disease, there is going

 

 22   to be a spillover benefit to the application of

 

 23   these to the processes themselves.

 

 24             So, this not a static, obviously a static

 

 25   situation, and I expect that the main driver for

 

                                                               287

 

  1   some of the new analytical techniques will not be

 

  2   process understanding, but rather will be

 

  3   understanding the biology, and that it is up to us

 

  4   to take those same techniques and those same

 

  5   methodologies and begin to apply them to the

 

  6   processes.

 

  7             The other piece of this, to emphasize a

 

  8   point I made earlier, by being able to do a lot of

 

  9   measurements on a small scale, one can take

 

 10   advantage of experimental design and look at your

 

 11   experimental space, so these techniques that allow

 

 12   you to do that are important.

 

 13             Another area, I mentioned doing

 

 14   large-scale fermentation type of experimental

 

 15   programs, but you need to do this for downstream,

 

 16   as well, and there is a fair amount of work, there

 

 17   is a modest, well, there is a little bit of work

 

 18   being done to miniaturize the downstream processes

 

 19   that hopefully should have the benefit of also

 

 20   being able to do design of experiments on a larger

 

 21   amount of downstream space at the same time, so

 

 22   there is yet another area of development

 

 23   particularly in the microfluidic space.

 

 24             DR. KIBBE:  Just a quick follow-up

 

 25   question of our experts over here.  One of the

 

                                                               288

 

  1   things I have noticed whenever we discuss PAT, is

 

  2   we deal with a tremendous amount of data influx, we

 

  3   get lots of data, and then we have to sort out the

 

  4   data that is really valuable to us.

 

  5             Is there a role to play for the

 

  6   ever-increasing power of the computational machine

 

  7   that sits next to the instrument?

 

  8             DR. COONEY:  Absolutely yes, not only in

 

  9   working your way through large data sets, but also

 

 10   learning how to do simulation both at the molecular

 

 11   scale and upwards.

 

 12             There is very interesting work being done

 

 13   with modeling of small molecule-protein

 

 14   interactions that is useful from a design point of

 

 15   view, but it is also useful to explain some of the

 

 16   phenomena that you see in a process.

 

 17             So, having large computational capacity is

 

 18   very important both from the passive data mining,

 

 19   as well as the proactive process simulation role.

 

 20             DR. KOCH:  I have to more than second

 

 21   that.  The number of sensors being developed, and

 

 22   that is just begging for a number sensor mining and

 

 23   then into the data mining, and then on into how do

 

 24   you handle the monstrous amounts of data.

 

 25             DR. KIBBE:  Just a personal opinion about

 

                                                               289

 

  1   accepting monstrous amounts of data is I sincerely

 

  2   hope that the companies analyze it, pick out what

 

  3   is important, and the FDA accepts only those things

 

  4   that are worth looking at, and doesn't demand every

 

  5   ton that comes through the door.

 

  6             DR. SINGPURWALLA:  Well, I have to

 

  7   disagree.  I am suspicious of data mining because

 

  8   you are looking at patterns.  You may look at

 

  9   patterns that are purely imaginary.  There is a

 

 10   classic example of consumption of alcohol and

 

 11   professor's salaries.  You know, there are dubious

 

 12   correlations that can come about.

 

 13             Now, having said that, I think when you

 

 14   are exploring any data--and I think you asked a

 

 15   very good question, and I am not sure if the

 

 16   question has been addressed--with a lot of data, we

 

 17   are collecting a lot of data, by itself, may not

 

 18   contain the knowledge of the information that you

 

 19   are really looking for.

 

 20             You may collect a lot of data which

 

 21   provides information which is not really relevant

 

 22   to what it is that you are interested in.  So, the

 

 23   whole idea is when you are doing a data analysis

 

 24   rather than data mining, what you have to do is

 

 25   have some kind of a hypothesis in mind, have some

 

                                                               290

 

  1   kind of a model in mind, and the model is never

 

  2   suggested by the data, the model is always

 

  3   suggested by the science that you are looking at

 

  4   and let the data then give you the unknowns of the

 

  5   particular model or help you change your model or

 

  6   help you update your model.

 

  7             So, I think your question is very nice and

 

  8   very important, and I think it goes back to why

 

  9   collect the data. You should have a purpose for

 

 10   collecting the data.  You should have an

 

 11   experimental design in mind when you collect the

 

 12   data, and the design itself should be driven by a

 

 13   certain hypothesis.  So, this is more of a

 

 14   philosophical comment.

 

 15             DR. KIBBE:  Off-line, we will talk about

 

 16   the philosophy of making observations about your

 

 17   surroundings and then developing the thesis and

 

 18   hypothesis versus having an hypothesis and making

 

 19   your observations fit it.

 

 20             Shall we go on to the next question?

 

 21             DR. COONEY:  I can't let this point go

 

 22   unnoticed. I think that you are absolutely right,

 

 23   and I like your hypothesis, and I think the

 

 24   experiment that has to be done to confirm it is to

 

 25   increase the salaries of professors.

 

                                                               291

 

  1             [Laughter.]

 

  2             DR. KIBBE:  What processes in biological

 

  3   manufacturing would benefit the most from

 

  4   implementation of PAT?  I think we are dealing with

 

  5   fermentation here, and the alcohol is just

 

  6   naturally connected somehow.  Go ahead.

 

  7             DR. COONEY:  I would address this question

 

  8   in two ways.  One is if you look at which

 

  9   particular products might benefit by early

 

 10   implementation, and I would suggest that the

 

 11   simpler the better, better to walk rather than run,

 

 12   taking very complex biological products made by

 

 13   very complex processes would be a very perhaps

 

 14   difficult place to begin, so that I think one needs

 

 15   to think about what are the logical targets.

 

 16             But then within the process, it is

 

 17   important to think about it, as well, because it's

 

 18   in the fermentation that you define the initial

 

 19   product that is being made, but a lot of the

 

 20   concerns about process variance occur once you have

 

 21   made the product and it is then subsequently being

 

 22   processed.

 

 23             So, that suggests that you need to

 

 24   methodically think through your entire process as

 

 25   you do for a drug substance, but because there are

 

                                                               292

 

  1   more steps, there is more complexity, obviously,

 

  2   there is more to do.

 

  3             But one of the characteristics of

 

  4   biologicals is that it is important to get the

 

  5   synthesis right, and then it is important to treat

 

  6   it right once it has been made.

 

  7             DR. DeLUCA:  Let me just add to that, I

 

  8   think it's a good follow-on, and I guess we have

 

  9   heard a lot about the biological process in

 

 10   fermentation, purification, and certainly we have a

 

 11   sensor technology in the Smart systems today to be

 

 12   able to handle that.

 

 13             I guess I wanted to move to the fill and

 

 14   finish end of it, and I guess I have a lot of

 

 15   questions.  You know, do you apply PAT to current

 

 16   products?  What properties can vary from unit to

 

 17   unit?  What does the variation mean in the

 

 18   pharmacological sense?  These are the types of

 

 19   questions.

 

 20             But with regards to the biological, it

 

 21   seems in the fill and finish that most of these

 

 22   freeze dried, so they are going to be lyophilized,

 

 23   and I think in lyophilization, this is not a

 

 24   trivial situation here, and I think you think that

 

 25   you put 10,000 vials into a chamber and you get out

 

                                                               293

 

  1   10,000 vials with little variation.

 

  2             You have to look at processing, and most

 

  3   products are processed and then filled in the

 

  4   containers.  In freeze drying, the processing takes

 

  5   place in an individual container.

 

  6             Each little container is processed after

 

  7   it's filled, and the heat that goes to that

 

  8   container is such that each vial doesn't see the

 

  9   same temperature, you like to have a small

 

 10   variation across the shelves in the type of flow,

 

 11   and the heating element, the fluid that goes

 

 12   through it to heat and freeze, but it isn't.

 

 13             So, you end up with, you could have

 

 14   product that has a variation in moisture, you can

 

 15   have products that vary in meltback collapse, so

 

 16   this can occur, so I think it is important that

 

 17   when we are looking at PAT and looking at the fill

 

 18   and finish, that moisture becomes very, very

 

 19   important in these lyophilized products and these

 

 20   biologicals, and I think applying things like NIR

 

 21   to that, I think makes this a very doable thing, to

 

 22   be able to do that with every product. It's a

 

 23   non-invasive procedure, and I think that is

 

 24   critical.

 

 25             So, you have to bring in robustness in

 

                                                               294

 

  1   here.  What can the product chemically tolerate in

 

  2   the way of moisture? I mean it is being freeze

 

  3   dried because obviously, it can't be put into a

 

  4   solution form, so moisture is going to have an

 

  5   effect.

 

  6             But what moisture content, can it tolerate

 

  7   5 percent or maybe it only can tolerate a half

 

  8   percent, or maybe there is an optimum moisture

 

  9   content that is good, because you are going to get

 

 10   into changes in tertiary and quaternary structure,

 

 11   aggregation, and whatnot with regards to moisture.

 

 12             So, I think that is an area that really

 

 13   lends itself to PAT, I think is in actually

 

 14   determining the moisture of these products, and

 

 15   again knowing where, you know, what kind of

 

 16   variation it can tolerate, and you have to somehow

 

 17   try to have some idea of the pharmacological effect

 

 18   of this, whatever the effect is of the moisture,

 

 19   does it really translate into a pharmacological

 

 20   effect, but I think that is an area that needs to

 

 21   be looked at.

 

 22             DR. KOCH:  I would certainly agree with

 

 23   that, but one other part of the biological drug

 

 24   manufacture, particularly fermentation, that I

 

 25   think needs to be addressed is just the

 

                                                               295

 

  1   fermentation itself in terms of reaction

 

  2   engineering.

 

  3             If you look at today's fermenters, they

 

  4   don't look that much different than they did 50, 60

 

  5   years ago.  Aeration is very important, as it

 

  6   nutrient and contact, so maybe the most effective

 

  7   fermentation is where you optimize those

 

  8   parameters, and huge vessels are not necessarily

 

  9   the way to do that.

 

 10             I think we are in a sunk capital situation

 

 11   where industry probably can't afford to redesign

 

 12   the approach, but there are some very interesting

 

 13   approaches, that if you could number up from the

 

 14   micro scale that Charles indicated, you might have

 

 15   a far more effective control of the material and

 

 16   far less of impurities that are being generated.

 

 17             DR. COONEY:  I would like to add another

 

 18   point to what Pat said.  When one is doing a

 

 19   de-bottlenecking exercise on a manufacturing

 

 20   process to try and improve the throughput, you

 

 21   begin that exercise from the end of the process and

 

 22   you work your way from the end forward.

 

 23             One very logical way of thinking about the

 

 24   application of the strategy, of PAT strategy, it is

 

 25   due to exactly the same thing, that if you

 

                                                               296

 

  1   understand, if you really understand the product

 

  2   and then you work your way back down the process,

 

  3   that makes it easier to de-bottleneck and design

 

  4   going forward.

 

  5             DR. KIBBE:  Since we started at the front,

 

  6   then, moving towards the back with the questions,

 

  7   we will keep going towards the back.

 

  8             The next question is for processes or

 

  9   products that do not currently allow direct product

 

 10   quality monitoring, what other strategies do you

 

 11   recommend for product quality control in addition

 

 12   to control of in-process parameters?

 

 13             DR. SINGPURWALLA:  I like this question.

 

 14             DR. KIBBE:  He likes this question.  It's

 

 15   a Bayesian question.

 

 16             DR. SINGPURWALLA:  Exactly.  I am really

 

 17   impressed with your insight and intuition.

 

 18             There is a technology, and there is a

 

 19   technology called information fusion.  Sometimes it

 

 20   is called information integration.  The basic idea

 

 21   is this.  The analogy here is like investigating a

 

 22   crime.  Some crime has been committed.  You don't

 

 23   know who has committed the crime. You are gathering

 

 24   all kinds of evidence, and then you are pooling

 

 25   that evidence in a very systematic way to make a

 

                                                               297

 

  1   probabilistic judgment about the crime.  You cannot

 

  2   make a judgment with certainty, because the only

 

  3   way to make a judgment to certainty is to see

 

  4   something.

 

  5             So, you have a similar situation here, and

 

  6   the problem you mention is common in other

 

  7   scenarios where you cannot directly observe the

 

  8   product.

 

  9             You are not allowed to either observe the

 

 10   product or test the product for whatever reason you

 

 11   have, but you have evidential information.  You

 

 12   have information on degradation, you have

 

 13   information on other kind of attributes, and how do

 

 14   you systematically integrate that information is a

 

 15   well-developed technology, and I think that would

 

 16   be germane here to the kind of question that you

 

 17   are raising.

 

 18             You have a process, you cannot directly

 

 19   observe it, but you presumably can observe other

 

 20   things related to it.  So, the question is how do

 

 21   you systematically pool that information, and there

 

 22   is a methodology, and, of course, it is Bayesian,

 

 23   as our chairman so wisely suggested, and it is

 

 24   available.

 

 25             DR. KIBBE:  Do you have a different

 

                                                               298

 

  1   method?

 

  2             DR. LAYLOFF:  No, I like Bayesian, but I

 

  3   was thinking that the monitoring and control, the

 

  4   in-process parameters may be polyvariate with

 

  5   respect to the quality of product.

 

  6             It may be a series of interactions on

 

  7   product quality, so the thing has to be linked

 

  8   together, so the evidentiary procedure may be many,

 

  9   many different mixtures of it to relate to the

 

 10   product quality.

 

 11             DR. SINGPURWALLA:  Just to add to that,

 

 12   you used the word polyvariate?

 

 13             DR. LAYLOFF:  No, I didn't.

 

 14             [Laughter.]

 

 15             DR. SINGPURWALLA:  It's multivariate.

 

 16             DR. LAYLOFF:  I know I did something

 

 17   wrong.

 

 18             DR. SINGPURWALLA:  The thing you want to

 

 19   be careful about is this multivariate information

 

 20   may be interdependent because the same phenomena

 

 21   can appear under two guises, so you don't want to

 

 22   add up, you know, the basic information should not

 

 23   be added up.  You have got to recognize the

 

 24   interdependence when there is a multivariate case,

 

 25   and therefore the technology, the mathematical

 

                                                               299

 

  1   technology that you need has to be nicely refined

 

  2   and carefully thought out, but the technology is

 

  3   what I am suggesting is purely an analytical

 

  4   technology, it is not a physical technology.

 

  5             DR. WEBBER:  Thank you.

 

  6             This question was mostly--I think that's a

 

  7   great answer, but we are looking at sort of, as Dr.

 

  8   Cooney pointed out, the oxygenation issues with

 

  9   fermentation, how that affects product.  If you can

 

 10   understand those, what sort of surrogates can you

 

 11   use to monitor or have a comfort level with the

 

 12   product quality based on looking at secondary

 

 13   parameters, but I think the answer you have given

 

 14   is one that we have to consider, as well, the

 

 15   analytical methods used to ensure that those aren't

 

 16   interfering with one another.

 

 17             DR. COONEY:  There is a fundamental

 

 18   problem with surrogates, and that is that many of

 

 19   them come from correlative observation, and the

 

 20   point was made quite appropriately earlier that

 

 21   when you take the data, develop a correlation, that

 

 22   may work within a certain amount of space with a

 

 23   certain set of assumptions, but it indeed is a

 

 24   correlation.

 

 25             I think the challenge that we have is to

 

                                                               300

 

  1   take that correlative knowledge and then create an

 

  2   hypothesis that, in fact, can be tested by one or

 

  3   more of the many techniques that we are talking

 

  4   about.

 

  5             I think what we are really talking about

 

  6   in this initiative is a change in the mind-set and

 

  7   the way that we think about developing and

 

  8   exploring and validating our processes, so there is

 

  9   going to be a lot of these iterations of learning,

 

 10   many of which will come from the surrogate

 

 11   procedures and correlative observations, but we

 

 12   need to drill down and understand that we are

 

 13   solving the right problem at the right time in the

 

 14   right way.

 

 15             DR. KIBBE:  Ajaz.

 

 16             DR. HUSSAIN:  I think the points are well

 

 17   made.  As we were putting the guidance together,

 

 18   one of the key aspects that we did say that in some

 

 19   cases, correlations would not be sufficient from a

 

 20   regulatory perspective, and we would look for

 

 21   causality as a means for making judgments, and so

 

 22   forth, and we sort of leaned that way, risk in a

 

 23   systems way, because keeping in mind, you are

 

 24   looking at a constrained space once it's in

 

 25   manufacturing.

 

                                                               301

 

  1             So, there are opportunities to utilize

 

  2   correlations in some low-risk areas, but also when

 

  3   there is a risk associated, you might prefer it to

 

  4   be causal rather than correlative.  That is how we

 

  5   sort of structured the guidance.

 

  6             DR. KOCH:  I guess this supports the

 

  7   Bayesian approach, as well, we will find out in a

 

  8   second, but rather than the crime analogy, I would

 

  9   like to think of something that is more like going

 

 10   to have a physical with a physician, where you are

 

 11   actually trying, in our product quality

 

 12   specifications, you know, assuming that is the

 

 13   perfect health you are looking for, but then

 

 14   develop a number of tests that would be analogous

 

 15   to doing body fluids or x-rays or a bunch of

 

 16   technologies, and then looking at the results that

 

 17   are coming back like in the physician's office, all

 

 18   of the tests are not going to be judged equally,

 

 19   but if you have a blood pressure and a lipid and an

 

 20   EKG, that is out of a predefined specification, you

 

 21   will start to spend your time at that first in

 

 22   order to see how the process is working.

 

 23             So, you will have a lot of data to work

 

 24   from, but you will have to make some assumptions

 

 25   early on in terms of what type of data relate more

 

                                                               302

 

  1   quickly to the final health of the product in this

 

  2   case.

 

  3             DR. KIBBE:  Jurgen, do you have any

 

  4   opinions so far?

 

  5             DR. VENITZ:  To respond I think to the top

 

  6   four or five questions in terms of how do you link

 

  7   all of this to the in-vitro potency, I think

 

  8   Patrick alluded to that a little bit when he talked

 

  9   about the fill stage, the late stage.  You are

 

 10   measuring attributes.  You may or may not know what

 

 11   they actually are other than they depend on some of

 

 12   your process variables.

 

 13             My question, as a pharmacologist, is

 

 14   always so, why should I care about that.  It is

 

 15   driven by your ability to measure, not necessarily

 

 16   by your ability to understand the consequences.

 

 17   You do know it is affected by your process, but you

 

 18   don't know whether it has any pharmacological

 

 19   consequences that I should care about.

 

 20             So, whenever you are looking at those

 

 21   steps, there has to be a linkage between whatever

 

 22   attributes you have to the ultimate pharmacological

 

 23   activity of the product.

 

 24             So, maybe that is my comment.

 

 25             DR. KIBBE:  Thank you, Jurgen.

 

                                                               303

 

  1             DR. WEBBER:  I certainly agree with that

 

  2   completely.  I think one can measure all sorts of

 

  3   things, but you have to look at what the critical

 

  4   product characteristics are, and that is something

 

  5   that has to be determined during the clinical

 

  6   development stage and product development stages.

 

  7             DR. VENITZ:  It goes beyond that.  I mean

 

  8   when I listen to the gentleman talk about what

 

  9   process analytical technology does, the way I

 

 10   understand, it is basically a statistical way of

 

 11   relating process variables and their impact, their

 

 12   criticality in terms of other attributes that you

 

 13   measure using some of those sensors that we heard

 

 14   about, but that doesn't tell me whether I should

 

 15   care about any of this, because you are really then

 

 16   changing your variables to affect your attributes

 

 17   in a way that you think it should be.

 

 18             But my question is, so what is your

 

 19   template, how do you know that that is the way your

 

 20   attributes should be, unless it is relevant to the

 

 21   pharmacologic activity of your product?

 

 22             If it is not relevant, then, yes, you

 

 23   might be improving your process, but it is

 

 24   cosmetic, it is not of any particular relevance for

 

 25   me to care about.  So, as part of this in the

 

                                                               304

 

  1   development stage, maybe not in manufacturing, the

 

  2   ultimate manufacturing stage, there has to be a

 

  3   linkage to the pharmacologic activity.

 

  4             DR. KIBBE:  Go ahead, Pat.

 

  5             DR. DeLUCA:  Just to add, I think it is

 

  6   critical that we have good control, I mean with

 

  7   regards to the fermentation before it gets to the

 

  8   fill and finish.  I think that is essential.  I

 

  9   think when the product gets to the point where it

 

 10   is going to be formulated and put into final dosage

 

 11   form, you want to have good control over that,

 

 12   because in that finishing, there could be a lot of

 

 13   variation.

 

 14             I just mentioned moisture before, but when

 

 15   you are freezing, you are freezing an amorphous

 

 16   form, and then when you are drying, you get a lot

 

 17   of conversion into the crystalline states, and

 

 18   there could be variation in the distribution of

 

 19   crystalline and amorphous form in the finished

 

 20   product, and that is going to affect the water

 

 21   content and the effects, so there is a lot of

 

 22   variation that can occur in that stage, that I

 

 23   think there needs to be control over.

 

 24             I support that there needs to be great

 

 25   control in the upstream processing, as you

 

                                                               305

 

  1   mentioned.

 

  2             DR. KIBBE:  Anybody else on the first

 

  3   four?

 

  4             That brings us to:  What additional

 

  5   elements should be incorporated in a training and

 

  6   certification program for reviewers and inspectors

 

  7   of biotechnology PAT?

 

  8             I am going to take the prerogative of the

 

  9   Chair to speak first, and then you all will tell me

 

 10   what mistakes I have made.

 

 11             I have been involved with educating

 

 12   college level students for years, and one of the

 

 13   things that I find is that they learn something in

 

 14   a specific incidence or example, but they don't

 

 15   learn things in the generalities, and one of the

 

 16   most powerful tools we have to handle all of this

 

 17   is a true and real understanding of the scientific

 

 18   method and the application of the scientific method

 

 19   to the problem in front of you, and not just

 

 20   learning how to do it in one situation, but

 

 21   learning how that works in any situation.

 

 22             I don't know whether you can get that into

 

 23   a training session, with a process that hasn't been

 

 24   well developed and isn't templated, when you look

 

 25   at the data you have in front of you and can use

 

                                                               306

 

  1   that.

 

  2             The second is critical thinking.  Often,

 

  3   students really don't know the difference between

 

  4   facts and opinions, because they have been trained

 

  5   over their life to accept someone who stands in

 

  6   front of them in a lecture hall and says something

 

  7   as if they were saying facts, when, in fact, most

 

  8   of the time we who lecture give opinions about

 

  9   everything and very little real facts.

 

 10             They also have a hard time differentiating

 

 11   results from conclusions.  They look at the result

 

 12   and immediately leap to a conclusion that isn't

 

 13   necessarily supported by the facts, and if we could

 

 14   add anything to anybody's training who are going to

 

 15   be involved in reviewing data or doing inspections,

 

 16   it is that level of sophistication that would be

 

 17   very helpful.

 

 18             DR. SINGPURWALLA:  I agree with you on

 

 19   that point certainly.  I really also think that

 

 20   there should be some kind of education and training

 

 21   on basically uncertainty, what is the meaning of

 

 22   uncertainty, how to quantify uncertainty, what are

 

 23   the different ways of quantifying uncertainty, what

 

 24   is the difference between variability and

 

 25   uncertainty, is there a difference, and basically,

 

                                                               307

 

  1   not statistics or statistical technology, but just

 

  2   the background of what it is all about, I think

 

  3   that would be very valuable because that seems to

 

  4   be running through completely all the way here.

 

  5             So, that is what I would like to add,

 

  6   "parochial."

 

  7             DR. LAYLOFF:  I think also it would be

 

  8   useful to turn to the biotechnology industry, to

 

  9   the people working in CMC, to try and help define

 

 10   what attributes reviewers and inspectors should

 

 11   have to properly evaluate, because those guys live

 

 12   with that stuff on a daily basis, and I am sure

 

 13   they would be willing to help.

 

 14             DR. KIBBE:  Judy.

 

 15             DR. BOEHLERT:  Finally, I am going to make

 

 16   a comment.  My background is in small molecules, so

 

 17   I am sorry, but it would seem to me with the

 

 18   complexity of these processes, that you might want

 

 19   to go to industry and sort of talk with them, and I

 

 20   think you know, as well, about what are the issues

 

 21   that can occur, because things go wrong in these

 

 22   processes that don't go wrong in conventional

 

 23   processes.

 

 24             You have adventitious contamination and

 

 25   things that don't happen elsewhere, so

 

                                                               308

 

  1   investigators, reviewers need to learn to ask the

 

  2   right questions and go beyond what they see to say,

 

  3   well, what about this, what about that, could this

 

  4   happen here, did this happen here, and that you

 

  5   need to train people to ask the right questions,

 

  6   because it's a whole different ball game when you

 

  7   get into these products.

 

  8             DR. KIBBE:  Anybody else?  Do we have

 

  9   anybody who hasn't spoken?  Would you like to

 

 10   comment on our discussion?

 

 11             DR. SELASSIE:  I think I will pass.

 

 12             DR. KIBBE:  What I am going to do now,

 

 13   unless Keith has something specific he needs us to

 

 14   do, is I am going to summarize.

 

 15             DR. WEBBER:  I didn't have anything

 

 16   specific for you to do now.  Maybe before you

 

 17   summarize, I would like to thank the committee

 

 18   certainly for getting together and addressing the

 

 19   issues and the questions that we have, giving

 

 20   presentations, and giving us your input on this

 

 21   difficult issue that we have ahead of us.

 

 22             DR. KIBBE:  You had five things that you

 

 23   wanted us to help you with and some of them we can

 

 24   help you with and sometime we will help you with.

 

 25             Starting with the first one, technology

 

                                                               309

 

  1   changes at an ever-increasing rate since the

 

  2   beginning of civilization, each breakthrough in

 

  3   technology is taking shorter and shorter periods of

 

  4   time.  if you wait two weeks, there will be a new

 

  5   technology to measure something.

 

  6             The question is what do you need to know,

 

  7   what good questions have you asked, and that is the

 

  8   core of the quality of scientific endeavor, so make

 

  9   sure you ask good questions, and there will be

 

 10   someone out there will develop a way of getting you

 

 11   an answer.

 

 12             No. 2, data collection is important.  I

 

 13   think there is fun in data mining, I enjoy it. I go

 

 14   looking for patterns and try to develop patterns,

 

 15   but the question really is are these patterns of

 

 16   correlation of cause and effect, how do you know

 

 17   the cause and effect, and this boils down to being

 

 18   able to think critically about the analytical data

 

 19   in front of you.

 

 20             No. 3, variability control seems to be the

 

 21   key.  If we know how much variation we can allow in

 

 22   any critical step in order to still maintain a good

 

 23   product, then, that is the variation we should

 

 24   allow, and we should really look at variability on

 

 25   each critical step in the process.

 

                                                               310

 

  1             If we know our critical steps, which is

 

  2   always an assumption that we make, and we hope we

 

  3   do, and we know the variability that will throw our

 

  4   process out of control, then, we know where we need

 

  5   to limit ourselves, and making intelligent choices

 

  6   about those limits are really important.

 

  7             Related observations.  I know my friend

 

  8   likes, under No. 4, Bayesian approach.  It all

 

  9   boils down to critical thinking about the things

 

 10   that you can measure and the things that you need

 

 11   to measure, just because you can measure it doesn't

 

 12   mean you need to know about it.  If it is something

 

 13   critical you need to measure, you need to find a

 

 14   way to measure it.

 

 15             This brings us to No. 5, which I think

 

 16   boils down to training people to think critically

 

 17   and to apply the scientific method appropriately.

 

 18   The quality of good science is the quality of the

 

 19   questions.

 

 20             The difference between a normal researcher

 

 21   and Albert Einstein is that the way he posed the

 

 22   questions allowed him to get breakthroughs and

 

 23   answers.  The other thing that he had that most of

 

 24   us don't have, and I won't say any one of you

 

 25   doesn't, is that he was never satisfied with the

 

                                                               311

 

  1   quality of the answer, and he always kept looking

 

  2   for better and better answers.

 

  3             Twain said that what we don't know doesn't

 

  4   get us in trouble.  It is what we know that ain't

 

  5   so.

 

  6             So, we have to be very careful to avoid

 

  7   thinking we know something about our process just

 

  8   because we made a measurement, and it really isn't

 

  9   something that describes the process, but it is

 

 10   just a convenient measurement.

 

 11             We find in clinical realm that often a

 

 12   technique comes along looking for a disease to

 

 13   diagnose, so don't look for a technique that

 

 14   diagnoses a disease you don't have or wouldn't even

 

 15   get.  Just look for the ones that help you get the

 

 16   answers.

 

 17             On that note, I will end whatever

 

 18   soliloquy I have.

 

 19             Helen, do you have a comment?  I saw you

 

 20   getting closer to the microphone.

 

 21             MS. WINKLE:  No.

 

 22             DR. KIBBE:  We are going to be back here

 

 23   at 8:30 tomorrow morning.

 

 24             [Whereupon, the meeting was recessed at

 

 25   4:30 p.m., to reconvene at 8:30 a.m., Wednesday,

 

                                                               312

 

  1   April 14, 2004.]

 

  2                              - - -